Next Article in Journal
Evaluation of Two Levels of Trace Mineral Supplementation to Beef Calves Prior to Weaning
Previous Article in Journal
Transcriptomic and Metabolomic Analyses Reveal the Attenuating Role of Cordycepin and Cordyceps militaris Extract on Acute Liver Injury Induced by LPS in Piglets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genome-Wide Association Study of Reproductive Traits in Large White Pigs

1
College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
2
State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
3
Yunfu Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
4
National Engineering Research Center for Breeding Swine Industry, WENS Foodstuff Group Co., Ltd., Yunfu 527400, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Animals 2024, 14(19), 2874; https://doi.org/10.3390/ani14192874
Submission received: 10 September 2024 / Revised: 2 October 2024 / Accepted: 5 October 2024 / Published: 6 October 2024
(This article belongs to the Section Animal Genetics and Genomics)

Abstract

:

Simple Summary

Understanding the genetics behind the reproductive traits of pigs is crucial for enhancing productivity and economic outcomes in the pork industry. In this study, we investigated the genetic factors influencing reproductive characteristics in Large White pigs. By analyzing the genetic data of 2237 sows from southern China, we identified specific genes and genetic markers associated with important reproductive traits, such as litter size. Our findings provide valuable insights that could help in developing targeted breeding strategies aimed at improving these traits. This research is not only significant for farmers and breeders but also contributes to a more sustainable and efficient approach to pig farming.

Abstract

(1) Background: Reproductive performance is crucial for the pork industry’s success. The Large White pig is central to this, yet the genetic factors influencing its reproductive traits are not well understood, highlighting the need for further research. (2) Methods: This study utilized Genome-Wide Association Studies to explore the genetic basis of reproductive traits in the Large White pig. We collected data from 2237 Large White sows across four breeding herds in southern China, focusing on eight reproductive traits. Statistical analyses included principal component analysis, linkage disequilibrium analysis, and univariate linear mixed models to identify significant single-nucleotide polymorphisms and candidate genes. (3) Results: Forty-five significantly related SNPs and 17 potential candidate genes associated with litter traits were identified. Individuals with the TT genotype at SNP rs341909772 showed an increase of 1.24 in the number of piglets born alive (NBA) and 1.25 in the number of healthy births (NHBs) compared with those with the CC genotype. (4) Conclusions: The SNPs and genes identified in this study offer insights into the genetics of reproductive traits in the Large White pig, potentially guiding the development of breeding strategies to improve litter size.

1. Introduction

Reproductive performance is a critical determinant of productivity and economic viability in the pork industry. Among various breeds, the Large White pig is renowned for its adaptability and reproductive performance, making it a staple in commercial pig farming globally. However, despite the breed’s importance, genetic factors underlying its reproductive traits remain incompletely understood [1]. The complexity of reproductive traits, which are influenced by a myriad of genetic and environmental factors, poses significant challenges in enhancing trait predictability and selection efficiency [2]. Particularly, litter size is a vital trait in the pork industry, directly impacting the economic outcomes of pig farming.
Reproductive traits in pigs, such as litter size, fertility, and piglet survival, are quintessential quantitative traits with low to moderate heritability [3]. This genetic architecture suggests that these traits are controlled by numerous genes with small effects rather than by a few major genes. Traditional phenotypic selection methods have achieved limited success, primarily due to the low heritability of these traits, which leads to slow genetic progress. Consequently, there is a pressing need for more precise and effective genetic improvement strategies.
In recent years, Genome-Wide Association Studies (GWASs) have emerged as a powerful tool for dissecting the genetic basis of complex traits in livestock. GWAS allows for the examination of the entire genome to identify associations between genetic markers and traits of interest. This approach has successfully identified numerous single-nucleotide polymorphisms (SNPs) and candidate genes associated with economically important traits in pigs, including coat color [4], meat quality [5], and reproductive performance [6]. Notably, GWAS on reproductive traits in pigs has revealed several loci and genes potentially affecting litter size and piglet viability, which are crucial for improving reproductive efficiency.
The present study aimed to conduct a comprehensive GWAS to identify SNPs and candidate genes associated with eight reproductive traits related to litter size in Large White pigs. The findings from this study are expected to contribute to the development of molecular breeding strategies that could expedite genetic improvements and, thus, enhance the productivity and profitability of the pork industry.

2. Materials and Methods

2.1. Animals and Traits

The animals used in this study were sourced from a swine breeding herd located in southern China, owned and managed by Wens Foodstuff Group Co., Ltd., based in Yunfu China. Data collection from these herds took place between November 2021 and May 2024. A total of 3368 records (total number born, TNB), only including first and second litters, were observed from 2237 Large White sows. The reproductive traits assessed included TNB, NBA, NHB, the rate of healthy births (rNHBs), the number of weak births (NWBs), the number of deformed fetuses (NDFs), the number of stillborn fetuses (NSBs), and the number of mummified pigs (MUMM). TNB represents the total number of piglets born in a single birthing event and is calculated as the sum of NBA, NSB, and MUMM. NBA is the total count of all surviving piglets, comprising NHB, NWB, and NDF. NHB denotes piglets with a birth weight of no less than 1.0 kg, whereas NWB refers to those piglets weighing less than 1.0 kg; however, neither NSB nor NWB exhibits genetic defects. The rNHB ratio is defined as the proportion of NHB to TNB. NDF pertains to piglets born with genetic anomalies, such as limb malformations or reproductive system disorders. NSB indicates the count of piglets that were stillborn or died during gestation. MUMM refers to cases where a pig fetus dies or undergoes degeneration after the 35th day of gestation without leading to a miscarriage. Ultimately, the eight traits, TNB, NBA, NHB, rNHB, NWB, NDF, NSB, and MUMM—were utilized for further analysis, with details provided in Table 1.

2.2. Genotypes and Quality Control

A total of 2237 tail tissue samples from Large White sows were collected, and genomic DNA was extracted using the QIAamp DNA Mini Kit (QIAGEN, Shanghai, China). The DNA quality was subsequently assessed via spectrophotometry NanoDropTM 2000 (Thermo Fisher Scientific, Waltham, MA, USA) and gel electrophoresis. Samples meeting the quality standards (minimum concentration of 50 ng/μL and an A260/A280 ratio between 1.8 and 2.0) were genotyped using the Affymetrix Wens 55K SNP Chip. Quality control measures for the genotyping data were implemented using Plink 1.90 [7], setting criteria such as a Hardy-Weinberg equilibrium threshold of 10−6, a minor allele frequency of 0.01, a minimum individual call rate of 0.95, and an SNP call rate of 0.99. Ultimately, 39,580 SNP markers met the quality control standards across all 2237 Large White pigs.

2.3. Statistical Analysis

Principal component analysis (PCA) was conducted using GCTA software version 1.93.2 beta [8], while linkage disequilibrium (LD) analysis was performed with PopLDdency software version 3.40 [9]. We analyzed the average LD decay distance across the whole genome where r2 = 0.2. Heritability was estimated using the GCTA software version 1.93.2 beta. The GWAS employed univariate linear mixed models (MLMs) via the GEMMA software version 0.98.5 [10], modeled as Y = Z β + W a + u + e , where Y represents the vector of phenotypic records, β is the vector of the corresponding coefficients including the intercept, a is the effect size of the marker, u is the vector of random effects, e is the vector of errors, Z is a matrix of covariates (fixed effect including farm-year-moth), and W is the vector of marker genotypes. The random effect u and errors e are assumed to follow multivariate normal distributions: ( u     M V N n   0 , λ τ 1 K ,   e     M V N n   ( 0 , τ 1 I n ) ), where λ is the ratio between the two variance components; K is a known relatedness matrix; I n is the identity matrix; and M V N n denotes the dimensional multivariate normal distribution. To establish thresholds for the genome-wide significance and suggestive significance, we based our calculation on 14,443 effectively independent SNP derived from independent markers and an LD block (defined as a set of SNPs with pairwise r2 values greater than 0.80). Consequently, the genome-wide significance threshold was set at 0.05/14,443 = 3.46 × 10−6, and the genome-wide suggestive significance threshold was set at 1/14,443 = 6.92 × 10−5. The GWAS results were visualized using the CMplot package in R version 4.3.2, generating Manhattan and quantile–quantile (Q-Q) plots. GWAS frequently identifies clusters of significant SNPs associated with a target trait within a putative region, likely due to the LD among them. To delineate the independence of all significant signals within such a region, conditional analyses were conducted using a mixed linear model (MLM), where the genotypes of peak SNPs were incorporated as covariates [11].

2.4. Identification of Candidate Genes and Analysis of Functional Enrichment

The physical locations of all SNPs and functional genes are mapped to the latest Sus scrofa 11.1 genome assembly, accessible at [https://ensembl.org/Sus_scrofa/Info/Index (accessed on 15 July 2024)]. Annotation was performed by identifying the nearest functional gene to each significant SNP, along with potential candidate genes selected based on their physiological and biochemical functions. Subsequent Gene Ontology (GO) term annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were conducted using KOBAS 3.0 [12]. Biological process GO terms and KEGG pathways were then selected based on an adjusted p-value threshold of <0.01.

3. Results and Discussion

Over recent decades, the selective breeding of Large White pigs has primarily concentrated on enhancing traits such as litter size to augment production efficiency and economic viability within the swine industry. Although this strategy has effectively increased yields, it has inadvertently escalated a range of reproductive challenges. These include NWB, NDF, NSB, and MUMM, thereby highlighting the intricate nature of reproductive traits governed by a complex interplay of genetic and environmental factors. The GWAS on reproductive traits in Large White pigs offers an exhaustive analysis aimed at elucidating the genetic underpinnings that influence these reproductive efficiencies and anomalies. By pinpointing specific single SNPs associated with both beneficial and detrimental reproductive outcomes, this research endeavors to establish a foundation for more holistic breeding programs. Such programs aim not only to enhance productivity but also to significantly improve the overall health and welfare of the animals, ensuring sustainable practices within the breeding sector.

3.1. Genetic Architecture and LD Decay

PCA depicted in Figure S1 reveals a homogenous genetic background among the pigs studied, indicating an absence of population stratification. This uniformity is crucial in GWAS as it suggests that any association detected between SNP markers and traits is less likely to be confounded by population structure. Further, LD decay analysis, illustrated in Figure S2, shows that LD decays to a correlation coefficient of 0.20 at about 800 kb. This LD decay distance is significant for GWAS as it informs the resolution at which genetic associations can be mapped. A shorter LD decay implies a higher resolution for locating genes associated with traits, whereas a longer LD decay suggests broader genomic regions harboring potential causative variants. In this case, the 800 kb LD decay provides a moderate resolution, facilitating the identification of candidate regions but requiring denser SNP panels or follow-up studies for precise gene mapping.

3.2. Heritability

In our study, heritability estimates for reproductive traits such as TNB, NBA, NHB, rNHB, NWB, NDF, NSB, and MUMM in both first and second litters ranged from 0.01 to 0.07, as detailed in Table 1. These low heritability values indicate that genetic factors make only a minimal contribution to the observed variance in these traits among pigs. Consequently, the limited genetic influence reflected by these heritabilities may restrict the effectiveness of GWAS in identifying significant genetic markers associated with these traits. This suggests that although genetic factors do influence these reproductive traits, their overall impact is modest. Many reports have also found these traits to have low heritability, generally ranging from 0.01 to 0.15 [13,14,15]. Therefore, larger sample sizes may be necessary to detect any substantial genetic contributions. Despite these challenges, we have still managed to identify some valuable GWAS findings. Moving forward, increasing the sample size to enhance the statistical power of GWAS is crucial for identifying GWAS findings related to litter size traits.

3.3. Genome’Wide Association Studies

The GWAS results are presented in Table 2 and Table 3 and Figure 1 and Figure 2. Specifically, Table 2 and Figure 1 display the GWAS results for the first-parity litter size trait, whereas Table 3 and Figure 2 focus on the GWAS results for the second-parity litter size trait. In total, 45 SNPs were identified across the traits TNB, NBA, NHB, rNHB, NWB, NDF, NSB, and MUMM, including two significant and 43 suggestive SNPs. Specifically, the GWAS results for the first parity identified 21 suggestive SNPs, while the results for the second parity revealed two significant and 23 suggestive SNPs. Q-Q plots were employed to demonstrate the potential inflation of p-values, as depicted in Figures S3 and S4. The average genomic inflation factors (λ) for litter traits ranged from 0.98 to 1.02, further indicating minimal population stratification.
The GWAS results for the first and second parity litter size traits reveal distinct genetic influences, as evidenced by the lack of shared quantitative trait loci (QTLs) between them. This disparity could be attributed to several factors. Firstly, genetic variation specific to each parity may be driven by different biological processes, such as maternal age, physiological changes, or environmental adaptations that occur between the first and second pregnancies [16]. Secondly, the reproductive performance in pigs, like many other traits, can be influenced by epigenetic modifications that differ from one parity to another. These modifications can affect gene expression without altering the DNA sequence, potentially leading to different genetic markers being identified in the GWAS of each parity [17]. Additionally, the interaction between genetic and environmental factors can vary across parities, further contributing to the unique genetic landscapes observed [18]. Lastly, the statistical power and the methodology used in identifying significant and suggestive SNPs might also differ slightly, affecting the detection of common QTLs between the two parities. This analysis underscores the complexity of genetic architecture in litter size traits and highlights the need for tailored strategies in genetic selection and management practices for different parities.

3.4. Candidate Genes for Litter Traits

An SNP, rs55618047, was significantly associated with TNB traits in the second parity, with the candidate gene PRMT6 identified in close proximity to this SNP. Research indicates that the PRMT6 gene may be associated with birth weight traits in pigs. A previous genome-wide association study (GWAS) identified the PRMT6 gene variant rs12097821, located at 1p13.3, as a genetic susceptibility locus for nonobstructive azoospermia (NOA) in the Han Chinese population [19]. Similarly, in a GWAS on Polish Holstein-Friesian bulls, PRMT6 was highlighted due to its proximity to significant SNPs—specifically rs109154964 and rs108965556 on chromosome 3—which are associated with variations in sperm concentration [20]. This gene has also been identified as a promising candidate for further research into the progression of geographic atrophy, a condition urgently requiring effective treatments [21]. Additionally, the PRMT6 variant rs2232016 has been recognized as a new susceptibility locus for gestational diabetes mellitus (GDM) in the Chinese Han population. In a case-control study involving North Macedonian men with idiopathic infertility, the rs12097821 variant in the PRMT6 gene was analyzed [22]. Although the study revealed no significant differences in genotype distributions between the infertile men and controls, a subsequent meta-analysis confirmed the association of rs12097821 with the risk of idiopathic male infertility [23]. These findings underscore the diverse roles of PRMT6 in reproductive traits and diseases, highlighting its importance in genetic research and potential therapeutic targets.
The SNP rs341909772 was significantly associated with both NBA traits in the second parity and NHB traits in the second parity, with the candidate gene COG6 identified near this SNP. Although there is no existing research linking the COG6 gene to reproductive or litter traits in pigs, it has been implicated in human health conditions. Specifically, the COG6 gene (rs7993214) was identified as a susceptibility locus for juvenile idiopathic arthritis in a study, which found an odds ratio (OR) of 0.76 and a p-value of (1.10 × 10−5), suggesting it may have a protective effect against the disease [24]. Additionally, while this gene was investigated as a potential genetic risk factor for psoriasis in the Chinese population, it did not demonstrate a significant association in this group, contrasting with findings from Western populations reported in other Genome-Wide Association Studies [25]. Furthermore, a GWAS identified a significant bone mineral density (BMD) locus on Chromosome 3, which includes the COG6 gene within a 300 Kbp region known to influence BMD. This locus coincides with a previously identified quantitative trait locus (QTL) for BMD, underscoring a potential role for COG6 in the regulation of bone density [26]. Individuals with the TT genotype at SNP rs341909772 demonstrated higher NBA (1.24) and NHB (1.25) values compared with those with the CC genotype and also exhibited higher NBA (0.45) and NHB (0.52) compared with those with the CT genotype (Figure 3). Although this pleiotropic SNP may not be the causal mutation, it has been shown to possess significant molecular breeding value in GWAS.
Two additional SNPs, rs3474737609 and rs81431697, have been significantly linked to NBA traits in the second parity, with the candidate genes OLFM3 and DOCK9 located nearby. In a GWAS of Colombian creole cattle breeds, Blanco Orejinegro and Sanmartinero, the OLFML3 gene emerged as a key player in reproductive traits, notably affecting the age at first calving and calving intervals [27]. Moreover, the proximity of OLFM3 to a newly identified Parkinson’s disease locus on chromosome 1p21 suggests its potential role in the genetic landscape of sporadic Parkinson’s disease [28]. This association may indicate a specific genetic influence on the development of the disease. The study also underscores the importance of the DOCK9 gene in the genetic profile of domestic pigs, indicating its influence on key phenotypic traits [29]. Alongside genes such as ACACA and MECR, DOCK9 is involved in the complex interaction between genotype and phenotype, particularly in traits relevant to domestication and breeding. Identified within a co-expression module enriched for genes implicated by bone mineral density GWAS, DOCK9 is highlighted as a potential causal genetic driver of BMD in humans [30]. It is located within the BMD GWAS loci, which has colocalizing eQTLs and exhibits altered BMD in mouse knockouts. Collectively, these findings enhance our understanding of the genetic factors that shape economic reproduction traits in livestock, highlighting the potential for targeted genetic improvements in breeding programs. Individuals possessing the GG genotype at SNP rs81431697 exhibited an average increase of 1.31 in NBA values compared with those with the AA genotype and a 0.60 increase compared with individuals with the GA genotype (Figure S5). Additionally, this SNP also possesses significant breeding value.
Moreover, SNP (rs55618047) was significantly associated with NHB traits in the first parity, with the candidate gene TXLNB located near this SNP. Three SNPs (rs341878379, rs1108768276, rs320524315) demonstrated significant associations with NHB traits in the second parity, with the candidate genes COL21A1 and ENSSSCG00000053606 identified near these SNPs. Furthermore, one SNP (rs81294332) showed a significant association with rNHB traits in the first parity, with the candidate gene SLC39A12 located in close proximity. Another SNP (rs320041489) was significantly associated with rNHB traits in the second parity, with the candidate gene MTHFSD identified near this SNP. TXLNB, which encodes a coiled-coil domain protein, has been identified as a critical regulator of cardiac proteostasis. It is capable of modulating protein synthesis, degradation, and autophagic processes in response to cardiac stress. Overexpression of TXLNB in neonatal rat cardiomyocytes modifies the typical hypertrophic response to phenylephrine by inhibiting the increase in cell size, highlighting its essential role in maintaining protein homeostasis during cardiac hypertrophy [31]. Research has indicated that specific causative mutations in the COL21A1 gene may play a pivotal role in the regulation of growth and carcass traits in pigs. These mutations in COL21A1 are potentially linked to critical biological processes that influence meat quality and yield [32]. GWAS have identified the SLC39A12 gene as significantly associated with sperm abnormalities, such as damaged cell necks and tails, in cryopreserved Holstein bull semen, suggesting its vital role in preserving the structural integrity of sperm during the cryopreservation process [33]. Additionally, real-time quantitative PCR was employed to validate a 496 kb copy number variation (CNV) region containing the MTHFSD gene on chromosome 6 in Xiang pigs, revealing significant copy number variations associated with reproductive traits. The presence of CNVs in the MTHFSD gene, which functions as an RNA-binding protein, has been shown to significantly affect litter size in Xiang pigs, indicating its crucial role in regulating mRNA metabolism and pig reproduction [34]. Further research has identified an SNP (rs336610321) in MTHFSD that is significantly associated with TNB in pigs [35]. Individuals carrying the GG genotype at SNP rs81294332 demonstrated an average rNHB value increase of 6.29 compared with those with the AA genotype and a 3.44 increase relative to carriers of the GA genotype (Figure S6). Therefore, this SNP also holds significant breeding value.
A total of 17 SNPs located within the 27.45–28.39 Mb region on chromosome 15 were significantly associated with NWB traits in the first parity. We conducted a conditional analysis based on the top SNP rs332278643, complemented by LD analysis. It was found that these SNPs exhibit a high level of linkage with the top SNP rs332278643, with r2 values exceeding 0.8. Additionally, the CNTNAP5 gene is located in proximity to this region (Figure 4). The CNTNAP5 gene, disrupted by a paternal microdeletion at 2q14.3 in an autism spectrum disorder (ASD) study, is implicated in neural communication and connectivity, as evidenced by its association with autism spectrum disorders and additional rare missense mutations found in extended analyses [36]. This gene’s role in neurodevelopmental processes highlights its potential impact on cognitive functions and behavioral traits associated with ASD. Eight SNPs were significantly associated with NSB traits in the second parity; of these, four SNPs were located on chromosome 6, three on chromosome 11, and one on chromosome 14. The most significant SNP, rs342135123, was found on chromosome 6, near the GSE1 gene. The GSE1 gene was identified as a novel locus significantly associated with tardive dyskinesia in schizophrenia patients in a Genome-Wide Association Study [37]. Additionally, we conducted a conditional analysis using the prominent SNP rs342135123, further supported by LD analysis (Figure S7). In this analysis, we included the genotypes of the key pleiotropic SNP rs342135123 as a covariate in the mixed linear model (MLM), which resulted in the p-value for the association with NDF dropping below the anticipated threshold. This analysis confirmed that these SNPs are tightly linked to the primary SNP rs342135123, with r2 values exceeding 0.8. Individuals carrying the GG genotype at SNP rs342135123 showed an average NDF value that was 0.81 lower compared with those with the CC genotype and 0.50 lower compared with carriers of the GC genotype (Figure S8). Thus, this SNP also demonstrates significant breeding value.
One SNP rs81310054 was significantly associated with NDF traits in the first parity, with PKD2 identified as the candidate gene. Eight SNPs located on chromosome 3 (from 60.15 Mb to 61.87 Mb) were significantly associated with NDF traits in the second parity, with SUCLG1 as the candidate gene. A conditional analysis was also conducted using the top SNP rs81304023, which was further supported by LD analysis (Figure S9). It was found that these SNPs are tightly linked to the primary SNP rs342135123, with r2 values exceeding 0.8. The PKD2 gene encodes Polycystin-2 (PC2), a protein that functions as a Ca2+-permeable nonselective cation channel, playing a crucial role in Ca2+ signaling and cellular Ca2+ homeostasis. Mutations in PKD2 are responsible for Autosomal Dominant Polycystic Kidney Disease (ADPKD), highlighting its significant role in renal epithelial cell function and health [38]. The SUCLG1 gene encodes succinate-CoA ligase, which plays a critical role in the mitochondrial energy metabolism pathway, and mutations in SUCLG1 are linked to significant clinical manifestations, including hepatopathy and hypertrophic cardiomyopathy [39]. One SNP rs321202796 was significantly associated with MUMM traits in the first parity, with KIAA0930 identified as the candidate gene. The KIAA0930 gene is implicated in influencing the cachexic phenotype in cancer cells, suggesting a role in cancer-associated wasting and metabolic dysregulation [40].

3.5. The Functional Enrichment Analysis for Litter Traits

The functional genes identified in this study are associated with the development of multiple tissues and the secretion of essential hormones during pregnancy, such as embryonic placenta development, peptidyl-arginine methylation, testis development, ovary development, sex hormone signaling, spermatogenesis, and oogenesis (Table S1). These factors are closely linked to the number of litters and the survival or mortality of piglets. Building on these results, future GWAS on swine reproductive traits should focus on these critical genetic markers for more precise breeding strategies. To effectively leverage these findings, it is advisable for breeding programs to consider incorporating these genetic markers to potentially enhance reproductive efficiency and outcomes, particularly in terms of litter size. This approach offers a practical framework that breeders could use to integrate genetic insights, thereby refining breeding strategies to improve reproductive traits.

4. Conclusions

In this study, we conducted a GWAS to identify SNPs and candidate genes associated with litter traits across the first two parities in the Large White pig population. We successfully detected 45 significantly related SNPs and 17 potential candidate genes, including COG6, GSE1, CNTNAP5, IRF8, PAN3, SLC39A12, SUCLG1, DOCK9, GFRA2, OLFM3, PRMT6, WASF3, KIAA0930, MTHFSD, PKD2, TXLNB, and COL21A1. These findings provide valuable tools for enhancing the quantity and quality of Large White litters through molecular marker-assisted or genomic selection. Additionally, this research deepens our understanding of the genetic basis of sow reproductive traits, facilitating targeted improvements and sustainable practices in swine breeding.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ani14192874/s1, Figure S1: PCA Plots for Large White Pig Population. Left: 2D PCA using PC1 and PC2. Right: 3D PCA using PC1, PC2, and PC3 to explore genetic diversity; Figure S2: LD Decay Plot for Large White Pig Population; Figure S3: Q-Q plots of GWAS with SNPs for litter traits at the first parity in the Large White population; Figure S4: Q-Q plots of GWAS with SNPs for litter traits at the second parity in the Large White population; Figure S5: The genotype effect plot of the SNP rs341909772, which is associated with NBA in the Large White population at the second parity, is displayed. (** p < 0.001); Figure S6: The genotype effect plot of the SNP rs81294332, which is associated with rNHB in the Large White population at the first parity, is displayed. (** p < 0.001); Figure S7: The regional association plot illustrates the primary signal (rs333336780) associated with NSB on SSC6 in the Large White population at the second parity; Figure S8: The genotype effect plot of the SNP rs342135123, which is associated with NSB in the Large White population at the second parity, is displayed. (** p < 0.001); Figure S9: The regional association plot illustrates the primary signal (rs81304023) associated with NDF on SSC3 in the Large White population at the second parity; Table S1: Significant Gene Ontology and KEGG Pathway Analyses for Eight Reproductive Traits.

Author Contributions

Conceptualization, C.T., C.S. and Z.W.; methodology, C.T. and C.S.; software, Y.H. and X.H.; validation, C.T., C.S. and Z.W.; formal analysis, Y.H., X.H. and Y.Z.; investigation, D.W., X.H. and C.T.; resources, C.T. and C.S.; data curation, Y.Z., Y.H., X.H. and D.W.; writing—original draft preparation, Y.H., X.H., C.T. and D.W.; writing—review and editing, Y.H., Y.Z., Z.W. and C.S.; visualization, C.T. and C.S.; supervision, C.T. and C.S.; project administration, Z.W.; funding acquisition, Z.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Natural Science Foundation of Guangdong Province, China, grant number 2018B030313011.

Institutional Review Board Statement

All procedures related to the animals in this study were performed in strict accordance with the guidelines for the care and use of experimental animals established by the Ministry of Science and Technology of the People’s Republic of China (Approval Number: 2006--398).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author due to the ongoing nature of the research project, which includes other unpublished work. This restriction is in place to protect the integrity and confidentiality of the ongoing research.

Acknowledgments

Many thanks to all the staff at the pig core breeding farms of Wens Foodstuff Group Co., Ltd. (Yunfu, China) for record collection.

Conflicts of Interest

Authors Xiaoyan He, Dan Wu, and Yuxing Zhang were employed by Wens Foodstuff Group Co., Ltd. This company provided the pigs used in this study from a swine breeding herd that it owns and manages. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Sell-Kubiak, E. Selection for litter size and litter birthweight in Large White pigs: Maximum, mean and variability of reproduction traits. Animal 2021, 15, 100352. [Google Scholar] [CrossRef]
  2. Knecht, D.; Środoń, S.; Duziński, K. The impact of season, parity and breed on selected reproductive performance parameters of sows. Arch. Anim. Breed. 2015, 58, 49–56. [Google Scholar] [CrossRef]
  3. Zhang, S.; Zhang, J.; Olasege, B.S.; Ma, P.; Qiu, X.; Gao, H.; Wang, C.; Wang, Y.; Zhang, Q.; Yang, H.; et al. Estimation of genetic parameters for reproductive traits in connectedness groups of Duroc, Landrace and Yorkshire pigs in China. J. Anim. Breed. Genet. 2020, 137, 211–222. [Google Scholar] [CrossRef] [PubMed]
  4. Schiavo, G.; Bovo, S.; Tinarelli, S.; Gallo, M.; Dall’Olio, S.; Fontanesi, L. Genome-wide association analyses for coat colour patterns in the autochthonous Nero Siciliano pig breed. Livest. Sci. 2020, 236, 104015. [Google Scholar] [CrossRef]
  5. Zhang, Y.; Zhang, J.; Gong, H.; Cui, L.; Zhang, W.; Ma, J.; Chen, C.; Ai, H.; Xiao, S.; Huang, L.; et al. Genetic correlation of fatty acid composition with growth, carcass, fat deposition and meat quality traits based on GWAS data in six pig populations. Meat Sci. 2018, 150, 47–55. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, J.; Wu, Z.; Chen, R.; Huang, Z.; Han, X.; Qiao, R.; Wang, K.; Yang, F.; Li, X.-J.; Li, X.-L. Identification of Genomic Regions and Candidate Genes for Litter Traits in French Large White Pigs Using Genome-Wide Association Studies. Animals 2022, 12, 1584. [Google Scholar] [CrossRef]
  7. Chang, C.C.; Chow, C.C.; Tellier, L.C.; Vattikuti, S.; Purcell, S.M.; Lee, J.J. Second-generation PLINK: Rising to the challenge of larger and richer datasets. GigaScience 2015, 4, 7. [Google Scholar] [CrossRef]
  8. Yang, J.; Lee, S.H.; Goddard, M.E.; Visscher, P.M. GCTA: A Tool for Genome-wide Complex Trait Analysis. Am. J. Hum. Genet. 2011, 88, 76–82. [Google Scholar] [CrossRef]
  9. Zhang, C.; Dong, S.-S.; Xu, J.-Y.; He, W.-M.; Yang, T.-L. PopLDdecay: A fast and effective tool for linkage disequilibrium decay analysis based on variant call format files. Bioinformatics 2019, 35, 1786–1788. [Google Scholar] [CrossRef] [PubMed]
  10. Zhou, X.; Stephens, M. Genome-wide efficient mixed-model analysis for association studies. Nat. Genet. 2012, 44, 821–824. [Google Scholar] [CrossRef] [PubMed]
  11. Genetic Investigation of ANthropometric Traits (GIANT) Consortium; Yang, J.; DIAbetes Genetics Replication and Meta-analysis (DIAGRAM) Consortium; Ferreira, T.; Morris, A.P.; Medland, S.E.; Madden, P.A.F.; Heath, A.C.; Martin, N.G.; Montgomery, G.W.; et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 2012, 44, 369–375. [Google Scholar] [CrossRef]
  12. Xie, C.; Mao, X.; Huang, J.; Ding, Y.; Wu, J.; Dong, S.; Kong, L.; Gao, G.; Li, C.-Y.; Wei, L. KOBAS 2.0: A web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res. 2011, 39 (Suppl. S2), W316–W322. [Google Scholar] [CrossRef] [PubMed]
  13. Ye, J.; Tan, C.; Hu, X.; Wang, A.; Wu, Z. Genetic parameters for reproductive traits at different parities in Large White pigs. J. Anim. Sci. 2018, 96, 1215–1220. [Google Scholar] [CrossRef] [PubMed]
  14. Oh, S.H.; Lee, D.H.; See, M.T. Estimation of Genetic Parameters for Reproductive Traits between First and Later Parities in Pig. Asian-Australas. J. Anim. Sci. 2005, 19, 7–12. [Google Scholar] [CrossRef]
  15. Yang, Y.; Gan, M.; Yang, X.; Zhu, P.; Luo, Y.; Liu, B.; Zhu, K.; Cheng, W.; Chen, L.; Zhao, Y.; et al. Estimation of genetic parameters of pig reproductive traits. Front. Vet. Sci. 2023, 10, 1172287. [Google Scholar] [CrossRef] [PubMed]
  16. Knol, E.F.; Nielsen, B.; Knap, P.W. Genomic selection in commercial pig breeding. Anim. Front. 2016, 6, 15–22. [Google Scholar] [CrossRef]
  17. Cameron, N.; Enser, M. Fatty Acid Composition of Lipid in Longissimus Dorsi Muscle of Duroc and British Landrace Pigs and Its Relationship with Eating Quality. Meat Sci. 1991, 29, 295–307. [Google Scholar] [CrossRef] [PubMed]
  18. Roehe, R.; Kennedy, B.W. Effect of Selection for Maternal and Direct Genetic Effects on Genetic Improvement of Litter Size in Swine. J. Anim. Sci. 1993, 71, 2891–2904. [Google Scholar] [CrossRef] [PubMed]
  19. Zou, S.; Li, Z.; Wang, Y.; Chen, T.; Song, P.; Chen, J.; He, X.; Xu, P.; Liang, M.; Luo, K.; et al. Association Study Between Polymorphisms of PRMT6, PEX10, SOX5, and Nonobstructive Azoospermia in the Han Chinese Population. Biol. Reprod. 2014, 90, 96. [Google Scholar] [CrossRef] [PubMed]
  20. Hering, D.; Olenski, K.; Kaminski, S. Genome-Wide Association Study for Sperm Concentration in Holstein-Friesian Bulls. Reprod. Domest. Anim. 2014, 49, 1008–1014. [Google Scholar] [CrossRef] [PubMed]
  21. Grassmann, F.; Harsch, S.; Brandl, C.; Kiel, C.; Nürnberg, P.; Toliat, M.R.; Fleckenstein, M.; Pfau, M.; Schmitz-Valckenberg, S.; Holz, F.G.; et al. Assessment of Novel Genome-Wide Significant Gene Loci and Lesion Growth in Geographic Atrophy Secondary to Age-Related Macular Degeneration. JAMA Ophthalmol 2019, 137, 867–876. [Google Scholar] [CrossRef] [PubMed]
  22. Yue, S.; Pei, L.; Lai, F.; Xiao, H.; Li, Z.; Zeng, R.; Chen, L.; Chen, W.; Liu, H.; Li, Y.; et al. Genome-wide analysis study of gestational diabetes mellitus and related pathogenic factors in a Chinese Han population. BMC Pregnancy Childbirth 2023, 23, 856. [Google Scholar] [CrossRef] [PubMed]
  23. Rajovski, S.; Vucic, N.; Karanovic, J.; Matijasevic, S.; Savic-Pavicevic, D.; Dobrijevic, Z.; Brajuskovic, G. Association of PRMT6, PEX10 and SOX5 genetic variants with idiopathic male infertility: Evidence from North Macedonian population and an updated meta-analysis. Genetika 2023, 55, 355–372. [Google Scholar] [CrossRef]
  24. Thompson, S.D.; Sudman, M.; Ramos, P.S.; Marion, M.C.; Ryan, M.; Tsoras, M.; Weiler, T.; Wagner, M.; Keddache, M.; Haas, J.P.; et al. The susceptibility loci juvenile idiopathic arthritis shares with other autoimmune diseases extend to PTPN2, COG6, and ANGPT1. Arthritis Rheum. 2010, 62, 3265–3276. [Google Scholar] [CrossRef] [PubMed]
  25. Li, X.; Yu, H.; Wu, G. Investigating the genetic association of HCP5, SPATA2, TNIP1, TNFAIP3 and COG6 with psoriasis in Chinese population. Int. J. Immunogenet. 2014, 41, 503–507. [Google Scholar] [CrossRef] [PubMed]
  26. Mesner, L.D.; Calabrese, G.M.; Al-Barghouthi, B.; Gatti, D.M.; Sundberg, J.P.; Churchill, G.A.; Godfrey, D.A.; Ackert-Bicknell, C.L.; Farber, C.R. Mouse genome-wide association and systems genetics identifies Lhfp as a regulator of bone mass. PLOS Genet. 2019, 15, e1008123. [Google Scholar] [CrossRef] [PubMed]
  27. De León, C.; Martínez, R.; Rocha, J.; Darghan, A. Research Article Selection of genomic regions and genes associated with adaptation and fertility traits in two Colombian creole cattle breeds. Evolution 2021, 20. [Google Scholar] [CrossRef]
  28. Hill-Burns, E.M.; Wissemann, W.T.; Hamza, T.H.; A Factor, S.; Zabetian, C.P.; Payami, H. Identification of a novel Parkinson’s disease locus via stratified genome-wide association study. BMC Genom. 2014, 15, 118. [Google Scholar] [CrossRef]
  29. Wang, K.; Wu, P.; Yang, Q.; Chen, D.; Zhou, J.; Jiang, A.; Ma, J.; Tang, Q.; Xiao, W.; Jiang, Y.; et al. Detection of Selection Signatures in Chinese Landrace and Yorkshire Pigs Based on Genotyping-by-Sequencing Data. Front. Genet. 2018, 9, 119. [Google Scholar] [CrossRef] [PubMed]
  30. Sabik, O.L.; Calabrese, G.M.; Taleghani, E.; Ackert-Bicknell, C.L.; Farber, C.R. Identification of a Core Module for Bone Mineral Density through the Integration of a Co-expression Network and GWAS Data. Cell Rep. 2020, 32, 108145. [Google Scholar] [CrossRef] [PubMed]
  31. McLendon, J.M.; Zhang, X.; Stein, C.; Abouassaly, G.; Witmer, N.; Boudreau, R.L. TXLNB is a Novel Regulator of Cardiac Proteostasis. FASEB J. 2018, 32, 864.11. [Google Scholar] [CrossRef]
  32. Blaj, I.; Tetens, J.; Bennewitz, J.; Thaller, G.; Falker-Gieske, C. Integrating imputed structural variants and tandem repeats in a GWAS for growth and carcass traits in F2 pig crosses. In Proceedings of the 12th World Congress on Genetics Applied to Livestock Production (WCGALP); Veerkamp, R.F., de Haas, Y., Eds.; Wageningen Academic Publishers: Wageningen, The Netherlands, 2022; p. 390. [Google Scholar]
  33. Dementieva, N.V.; Dysin, A.P.; Shcherbakov, Y.S.; Nikitkina, E.V.; Musidray, A.A.; Petrova, A.V.; Mitrofanova, O.V.; Plemyashov, K.V.; Azovtseva, A.I.; Griffin, D.K.; et al. Risk of Sperm Disorders and Impaired Fertility in Frozen–Thawed Bull Semen: A Genome-Wide Association Study. Animals 2024, 14, 251. [Google Scholar] [CrossRef]
  34. Ran, X.; Pan, H.; Huang, S.; Liu, C.; Niu, X.; Li, S.; Wang, J. Copy number variations of MTHFSD gene across pig breeds and its association with litter size traits in Chinese indigenous Xiang pig. J. Anim. Physiol. Anim. Nutr. 2018, 102, 1320–1327. [Google Scholar] [CrossRef] [PubMed]
  35. Balogh, E.E.; Gábor, G.; Bodó, S.; Rózsa, L.; Rátky, J.; Zsolnai, A.; Anton, I. Effect of single-nucleotide polymorphisms on specific reproduction parameters in Hungarian Large White sows. Acta Vet. Hung. 2019, 67, 256–273. [Google Scholar] [CrossRef] [PubMed]
  36. Pagnamenta, A.T.; Bacchelli, E.; de Jonge, M.V.; Mirza, G.; Scerri, T.S.; Minopoli, F.; Chiocchetti, A.; Ludwig, K.U.; Hoffmann, P.; Paracchini, S.; et al. Characterization of a Family with Rare Deletions in CNTNAP5 and DOCK4 Suggests Novel Risk Loci for Autism and Dyslexia. Biol. Psychiatry 2010, 68, 320–328. [Google Scholar] [CrossRef] [PubMed]
  37. Lim, K.; Lam, M.; Zai, C.; Tay, J.; Karlsson, N.; Deshpande, S.N.; Thelma, B.K.; Ozaki, N.; Inada, T.; Sim, K.; et al. Genome wide study of tardive dyskinesia in schizophrenia. Transl. Psychiatry 2021, 11, 351. [Google Scholar] [CrossRef]
  38. Cantero, M.d.R.; Cantiello, H.F. Polycystin-2 (TRPP2): Ion channel properties and regulation. Gene 2022, 827, 146313. [Google Scholar] [CrossRef] [PubMed]
  39. Carrozzo, R.; Verrigni, D.; Rasmussen, M.; de Coo, R.; Amartino, H.; Bianchi, M.; Buhas, D.; Mesli, S.; Naess, K.; Born, A.P.; et al. Succinate-CoA ligase deficiency due to mutations in SUCLA2 and SUCLG1: Phenotype and genotype correlations in 71 patients. J. Inherit. Metab. Dis. 2016, 39, 243–252. [Google Scholar] [CrossRef]
  40. Zuo, Y.; Zhang, L.; Tang, W.; Tang, W. Identification of prognosis-related alternative splicing events in kidney renal clear cell carcinoma. J. Cell. Mol. Med. 2019, 23, 7762–7772. [Google Scholar] [CrossRef]
Figure 1. Manhattan plots of GWAS with SNPs for litter traits at the first parity in the Large White populations. In the Manhattan plots, the yellow and red lines represent the 5% genome-wide and chromosome-wide (suggestive) Bonferroni-corrected thresholds, respectively.
Figure 1. Manhattan plots of GWAS with SNPs for litter traits at the first parity in the Large White populations. In the Manhattan plots, the yellow and red lines represent the 5% genome-wide and chromosome-wide (suggestive) Bonferroni-corrected thresholds, respectively.
Animals 14 02874 g001
Figure 2. Manhattan plots of GWAS with SNPs for litter traits at the second parity in the Large White populations. In the Manhattan plots, the yellow and red lines represent the 5% genome-wide and chromosome-wide (suggestive) Bonferroni-corrected thresholds, respectively.
Figure 2. Manhattan plots of GWAS with SNPs for litter traits at the second parity in the Large White populations. In the Manhattan plots, the yellow and red lines represent the 5% genome-wide and chromosome-wide (suggestive) Bonferroni-corrected thresholds, respectively.
Animals 14 02874 g002
Figure 3. The genotype effect plot of the pleiotropic SNP rs341909772, which is associated with both NBA and NHB in the Large White population at the second parity, is displayed. On the left, the plot shows the effect of SNP rs341909772 on NBA, and on the right, the effect on NHB (** p < 0.001).
Figure 3. The genotype effect plot of the pleiotropic SNP rs341909772, which is associated with both NBA and NHB in the Large White population at the second parity, is displayed. On the left, the plot shows the effect of SNP rs341909772 on NBA, and on the right, the effect on NHB (** p < 0.001).
Animals 14 02874 g003
Figure 4. The regional association plot illustrates the primary signal (rs332278643) associated with NWB on SSC15 in the Large White population at the second parity. The plot displays negative log10 p-values of SNPs (y-axis) according to their chromosomal positions (x-axis). The red line indicates the genome-wide significance level. The primary SNPs are marked by red triangles. The left and right panels of the figure show the association results for NWB before and after conditional analysis on rs332278643, respectively. The color of the dots in the plot represents the level of linkage disequilibrium (LD) with the primary signal (rs332278643), where redder dots indicate higher LD levels, and yellower dots indicate lower LD levels.
Figure 4. The regional association plot illustrates the primary signal (rs332278643) associated with NWB on SSC15 in the Large White population at the second parity. The plot displays negative log10 p-values of SNPs (y-axis) according to their chromosomal positions (x-axis). The red line indicates the genome-wide significance level. The primary SNPs are marked by red triangles. The left and right panels of the figure show the association results for NWB before and after conditional analysis on rs332278643, respectively. The color of the dots in the plot represents the level of linkage disequilibrium (LD) with the primary signal (rs332278643), where redder dots indicate higher LD levels, and yellower dots indicate lower LD levels.
Animals 14 02874 g004
Table 1. Summary statistics of litter traits in Large White populations.
Table 1. Summary statistics of litter traits in Large White populations.
ParityTraitNMeanSDMinimumMaximumHeritability
1TNB203617.563.596250.04
NBA195514.723.665230.02
NHB195813.613.504210.04
rNHB197877.2716.60151000.02
NWB19910.891.34050.01
NDF20220.020.15010.01
NSB19371.881.66060.01
MUMM19140.661.07040.01
2TNB133218.533.346250.04
NBA130615.593.645230.06
NHB130614.143.274210.05
rNHB130876.6414.47151000.01
NWB12981.281.45050.01
NDF13280.020.15010.01
NSB12581.981.69060.01
MUMM12920.611.02040.01
TNB, Total number born; NBA, number of piglets born alive; NHB, number of healthy births; rNHB, the rate of NHB; NWB, number of weak births; NDF, number of deformed fetuses; NSB, number of stillborn; MUMM, mummified pig.
Table 2. Summary of GWAS results for first-litter size trait in the Large White pig population.
Table 2. Summary of GWAS results for first-litter size trait in the Large White pig population.
TraitSSCrsPositionAlleleMafpCandidate Gene
NHB1rs5561804725284574C/T0.3153156.05448 × 10−5TXLNB
rNHB10rs8129433244308657A/G0.1804712.21049 × 10−5SLC39A12
NWB15rs33640196427459405T/C0.2295842.02398 × 10−5CNTNAP5
NWB15rs33356963627509302T/C0.2294392.02405 × 10−5CNTNAP5
NWB15rs8130416827531806A/G0.2186869.90197 × 10−6CNTNAP5
NWB15rs33597313327581413C/T0.2215931.69344 × 10−5CNTNAP5
NWB15rs33366509327738575A/G0.2208668.04687 × 10−6CNTNAP5
NWB15rs34336877627746996C/G0.2210119.97989 × 10−6CNTNAP5
NWB15rs32061930427755517A/G0.2208668.04687 × 10−6CNTNAP5
NWB15rs8130559927775994A/G0.2208668.04687 × 10−6CNTNAP5
NWB15rs33743259127785758A/G0.2215931.14719 × 10−5CNTNAP5
NWB15rs32797603227883239A/T0.2205756.59095 × 10−6CNTNAP5
NWB15rs33227864327892641T/G0.2205756.57329 × 10−6CNTNAP5
NWB15rs8147887228021915T/G0.2202856.82459 × 10−6CNTNAP5
NWB15rs347095822028183106T/C0.2198497.14878 × 10−6CNTNAP5
NWB15rs32713514028279474G/T0.2199947.4039 × 10−6CNTNAP5
NWB15rs8145214128293171A/C0.2199947.4039 × 10−6CNTNAP5
NWB15rs32258204928347013A/G0.2199947.4039 × 10−6CNTNAP5
NWB15rs33876024528395070T/C0.2197046.6822 × 10−6CNTNAP5
NDF8rs81310054131016420A/C0.1049115.92047 × 10−5PKD2
MUMM5rs3212027964183089T/C0.1213315.00831 × 10−5KIAA0930
SSC, Sus scrofa chromosome; TNB, Total number born; NBA, number of piglets born alive; NHB, number of healthy births; rNHB, the rate of NHB; NWB, number of weak births; NDF, number of deformed fetuses; NSB, number of stillborn; MUMM, mummified pig.
Table 3. Summary of GWAS results for second-litter size trait in the Large White pig population.
Table 3. Summary of GWAS results for second-litter size trait in the Large White pig population.
TraitSSCrsPositionAlleleMAFpGenes
TNB4rs344560170113119104C/T0.3872424.308 × 10−5PRMT6
NBA4rs3474737609116388595A/T0.09561174.258 × 10−5OLFM3
NBA11rs34190977214909631T/C0.4478357.613 × 10−6COG6
NBA11rs8143169767830653G/A0.3728573.524 × 10−5DOCK9
NHB7rs34187837929614695A/C0.4064236.299 × 10−5COL21A1
NHB11rs34190977214909631T/C0.4478351.759 × 10−6COG6
NHB15rs11087682766547063C/A0.4091836.827 × 10−5ENSSSCG00000053606
NHB15rs3205243156384510C/T0.3437955.407 × 10−5ENSSSCG00000053606
rNHB6rs3200414892728087T/C0.4026456.856 × 10−5MTHFSD
NSB6rs3333367802814834C/T0.2746291.398 × 10−5IRF8
NSB6rs3347646152387727T/C0.3218545.853 × 10−5MTHFSD
NSB6rs3367296842454772T/C0.2736126.613 × 10−5MTHFSD
NSB6rs3421351233342934G/C0.3807032.223 × 10−6GSE1
NSB11rs3294674105544200C/T0.1069466.536 × 10−5PAN3
NSB11rs3458958365600114C/T0.09982561.604 × 10−5PAN3
NSB11rs814308594296303C/T0.1402214.744 × 10−5WASF3
NSB14rs3379192495225585C/T0.08936364.035 × 10−5GFRA2
NDF3rs8130402360157640T/C0.08384192.389 × 10−5SUCLG1
NDF3rs8133064760211109T/C0.08384192.389 × 10−5SUCLG1
NDF3rs8132703060216589T/C0.08384192.389 × 10−5SUCLG1
NDF3rs33224467960266762G/A0.08384192.389 × 10−5SUCLG1
NDF3rs8129981160914644C/G0.08413252.392 × 10−5SUCLG1
NDF3rs33679980161636339C/T0.08427782.743 × 10−5SUCLG1
NDF3rs8124450461851017C/T0.08442312.541 × 10−5SUCLG1
NDF3rs8132605061878711A/G0.08398722.391 × 10−5SUCLG1
SSC, Sus scrofa chromosome; TNB, Total number born; NBA, number of piglets born alive; NHB, number of healthy births; rNHB, the rate of NHB; NWB, number of weak births; NDF, number of deformed fetuses; NSB, number of stillborn; MUMM, mummified pig.
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

Hong, Y.; Tan, C.; He, X.; Wu, D.; Zhang, Y.; Song, C.; Wu, Z. Genome-Wide Association Study of Reproductive Traits in Large White Pigs. Animals 2024, 14, 2874. https://doi.org/10.3390/ani14192874

AMA Style

Hong Y, Tan C, He X, Wu D, Zhang Y, Song C, Wu Z. Genome-Wide Association Study of Reproductive Traits in Large White Pigs. Animals. 2024; 14(19):2874. https://doi.org/10.3390/ani14192874

Chicago/Turabian Style

Hong, Yifeng, Cheng Tan, Xiaoyan He, Dan Wu, Yuxing Zhang, Changxu Song, and Zhenfang Wu. 2024. "Genome-Wide Association Study of Reproductive Traits in Large White Pigs" Animals 14, no. 19: 2874. https://doi.org/10.3390/ani14192874

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