Go to Top Go to Bottom
Anim Biosci > Volume 36(6); 2023 > Article
Luan, Ruan, Qiu, Ye, Zhou, Yang, Sun, Ma, Wu, Yang, Yang, Zheng, Cai, and Huang: Genome-wide association study for loin muscle area of commercial crossbred pigs

Abstract

Objective

Loin muscle area (LMA) is an important target trait of pig breeding. This study aimed to identify single nucleotide polymorphisms (SNPs) and genes associated with LMA in the Duroc×(Landrace×Yorkshire) crossbred pigs (DLY).

Methods

A genome-wide association study was performed using the Illumina 50K chip to map the genetic marker and genes associated with LMA in 511 DLY pigs (255 boars and 256 sows).

Results

After quality control, we detected 35,426 SNPs, including six SNPs significantly associated with LMA in pigs, with MARC0094338 and ASGA0072817 being the two key SNPs responsible for 1.77% and 2.48% of the phenotypic variance of LMA, respectively. Based on previous research, we determined two candidate genes (growth hormone receptor [GHR] and 3-oxoacid Co A-transferase 1 [OXCT1]) that are associated with fat deposition and muscle growth and found further additional genes (MYOCD, ARHGAP44, ELAC2, MAP2K4, FBXO4, FBLL1, RARS1, SLIT3, and RANK3) that are presumed to have an effect on LMA.

Conclusion

This study contributes to the identification of the mutation that underlies quantitative trait loci associated with LMA and to future pig breeding programs based on marker-assisted selection. Further studies are needed to elucidate the role of the identified candidate genes in the physiological processes involved in LMA regulation.

INTRODUCTION

Pork is an important source of animal protein for humans, and the lean meat percentage is the most important breeding goal in important economic traits [1]. The loin muscle area (LMA), which is referred for the measurement of the large muscles on the back of the pig, also mainly constitutes the meat in the pork chop [2]. The LMA has a strong genetic correlation with lean meat percentage and is therefore valuable for predicting lean meat percentage [3]. The LMA is also helpful in predicting carcass traits, artificial selection, and growth experiments [4]. The key factors affecting psoas muscle growth mainly include nutrition (such as the protein content of the diet), management (such as the environment of the pig farm), and genetics. According to previous studies, the heritability of LMA is estimated to be between 0.35 to 0.47 [57], which is a moderately heritable trait and can be genetically improved.
In actual production, the most common way to measure LMA is through an A scan, however due to the error of the machine itself and other reasons it is difficult to get an accurate measurement [8]. Subsequent slaughter measurements of the LMA greatly reduces the error. But difficulties in phenotypic measurement still hinder the selection of LMA. With the advent of molecular breeding, in order to dissect the genetic molecular mechanisms of LMA, researchers have used different strategies to study different populations; for instance, the exploration of microsatellite markers in the pig genome and genome scans to detect quantitative trait loci (QTLs) in LMA [9] or using molecular genome scan analysis to identify chromosomal regions influencing LMA in pigs [9]. In the pig QTLs database [10] (Release 48, Aug 24, 2022), a total of 35,846 QTLs are identified, of which 416 are associated with LMA (1.16% of 35,846 QTLs), but the number of QTLs is still relatively low. QTL mapping throughout the genome for linkage mapping is normally difficult due to the low marker density, and most QTLs are usually maintained at large confidence intervals (over 10 Mb, including hundreds of genes), which severely hinders the optimization of plausible candidate genes [11]. Despite some progress in traditional genetic improvement of carcass and growth traits, elucidation of the biological mechanisms of LMA is still difficult and challenging [12].
With the rapid advancement of the livestock and poultry genome project, high-throughput pig single nucleotide polymorphism (SNP) chips were commercialized, which could effectively improve the accuracy of gene mapping in the pig genome [13]. Genome-wide association analysis (GWAS) has been widely used in humans as a powerful strategy to detect genetic variants associated with complex traits. In the studies on pigs, GWAS has gradually become an indispensable tool for exploring the genetic variation and structure of major economically advantageous traits in pigs [1416]. GWAS was also used to detect the underlying genetic mechanism for LMA. He et al [17] used the Laiwu pig population for GWAS analysis and identified three candidate genes (far upstream element binding protein 3 [FUBP3], myosin heavy chain [MYH] family, leucine-rich repeats and guanylate kinase domain containing [LRGUK]). Ding et al [18] performed a genome-wide analysis for copy number variation (CNV) detection using GeneSeek Porcine SNP50 Bead chip data of 3,770 pigs and identified one CNV region (CNVR) associated with LMA. Fan et al [19], used the IIuminaPorcineSNP60K chip to perform GWAS analysis on 820 female commercial pigs from Large White×Landrace cross and found two significant SNPs, M1GA0002180 and M1GA0002244, and identified the insulin-like growth factor 2 (IGF2) gene as a candidate gene. Zhuang et al [20] performed meta-GWAS of 6043 Duroc pigs for LMA and loin muscle depth traits and identified eight potential candidate genes. However, although previous studies have been helpful to LMA research, the key genes that influence LMA are yet to be identified, and the results of GWAS on LMA of pigs are inconsistent; consequently, the genetic mechanism of LMA requires further study. Furthermore, as a favorite source of pork, research on the LMA of DLY pigs is even more necessary. Therefore, we used LMA measurements of 511 DLY slaughtered pigs to perform GWAS to find important molecular markers and candidate genes for LMA. We hoped to provide a theoretical basis to explain the genetic basis of LMA and the development of molecular breeding, as well as provide biological guidance for subsequent breeding practices.

MATERIALS AND METHODS

Ethics approval

Treatment of all animals used in this study was as per the guidelines for the care and use of experimental animals established by the Ministry of Agriculture of China. Tissue samples from pigs were collected after obtaining approval from the ethics committee of South China Agricultural University (Guangzhou, China; approval no 2018F098).

Experimental animals

In this study, 511 DLY pigs (255 boars and 256 sows) raised under the same rearing standards were obtained from Wen’s Food Group Co., Ltd (Guangdong, China). Measurement of the LMA was carried out after slaughter. Specifically, the longissimus dorsi muscle was sampled at the last rib, and a sulfuric acid paper was used to draw the eye muscle area, and then a planimeter or a square grid was used. The area drawn on the sulfate paper was measured to determine LMA.

Genotyping and quality control

DNA was extracted from pig ear tissue using the phenol-chloroform method. DNA quality was measured by electrophoresis, and the absorption ratio (A260/280) was estimated. All quality DNA samples were diluted to 50 ng/μL, as described by Ding et al [21]. Genotyping was performed using GeneSeek porcine 50 K SNP chip from Illumina, then PLINKv1.09 [22] software was used for data editing. The quality control (QC) conditions for individuals were SNP markers call rate <95%, the QC conditions for SNPs were: minor allele frequencies <1%, and Hardy–Weinberg test p<10−06 were excluded. Unmapped SNPs and SNPs located on sex chromosomes were removed. Finally, after QC, a total of 35,426 SNPs were used for subsequent GWAS on LMA.

Single-locus genome-wide association study

The GWAS was performed using the method proposed by Zhou and Stephens [23]. To correct the population structure, an efficient mixed linear model was used to analyze the effect of a single SNP on phenotypic traits to develop the genetic algorithm [24]. GEMMA v0.94.1 software was used for association analysis of the characteristics [23], and the statistical formula is as follows:
y=Wα+xβ+μ+ɛu~MVNn(0,λτ^(-1)K),ɛ~MVNn(0,τ^(-1)In
where y is the vector of phenotypic values for LMA for all individuals and W is the correlation matrix based on appropriate fixed effects. To ensure that we could choose suitable fixed effects, we used EIGENSTRAT [25] to test the significance of the top five principal components (PCs) as covariables, and conducted analysis of variance by R language for the fixed effects of the selection. According to the test results (Supplementary Table S1), we finally determined five eigenvectors of principal components, slaughter batch and live weight were included as fixed affects. α is the vector of the corresponding coefficients, including the intercept, x is the vector of SNP marker genotypes, β is the corresponding effective vector of SNP markers, u and ɛ are vectors of random effects and random errors, respectively, τ^(−1) is the variance of the random errors. λ is the ratio between the two variance components, K is the standard correlation matrix of software estimation, I is an identity matrix, and MVN is a multivariate normal distribution. The heritability of LMA was estimated by the genome-wide complex trait analysis (GCTA) software.

Identification of significant single-nucleotide polymorphisms associated with loin muscle area

Significant SNPs were identified for LMA as those that surpassed the threshold with a false discovery rate (FDR) controlled at 0.01 [26]. The threshold p-value was estimated as follows:
p=FDR×n/m
where n represents the number of SNPs with p<0.01 in the GWAS results, ordered ascendingly by their effects, and m is the number of analyzed SNPs. In brief, the final selected threshold was 1.13×10−04, and the obtained GWAS results were visualized using R language [27]. The phenotypic variance explained by each significant SNP was estimated by the GCTA v1.94 software with “--reml” parameter [28,29].
Candidate genes 0.5 Mb upstream and downstream of significant SNPs were screened, and the functional genes were annotated on the Sus scrofa 11.1 genome from Ensemble genome database v107 (http://asia.ensembl.org/Susscrofa/Info/Index). The significant annotated genes were selected to conduct Kyoto encyclopedia of genes and genomes (KEGG) pathways and gene ontology (GO) analyses by using the KOBAS3.0 database [30]. The significance of the enriched terms was assessed using Fisher’s exact test, with p<0.05, and the genes involved in biological processes were detected. In addition, the NCBI and Genecard database were used to query gene functions and determine promising candidates.

Quantitative trait loci enrichment analysis

We used a R package called GALLO [31,32] to perform QTL enrichment. We perform QTL enrichment analysis by using our significant SNPs about LMA in order to further explore the influence of these SNPs on LMA.

RESULTS

Phenotype analysis and heritability estimation

After QC, 35,426 SNPs with genotypes on 511 DLY pigs were finally retained for subsequent analyses. The descriptive statistics of LMA for the 511 pigs are listed in Table 1, according to which, the average was 73.82±9.33 (cm2). The univariate model was used to estimate the SNP-based heritability of the LMA trait. The phenotypic variance was categorized into the variance, explained by the genetic component and the residual variance comprising the genetic relationship matrix. The genome heritability of LMA was 0.35±0.096, indicating moderate heritability. The results were similar to those published earlier, suggesting the accuracy of our slaughter phenotype determination and that the genetic technology could effectively promote the genetic improvement of LMA.

Single-locus genome-wide association studies for loin muscle area

After QC using PLINK software, we obtained a total of 35,426 SNPs for subsequent analysis; then GEMMA software was used for GWAS analysis. Because Bonferroni correction is too conserved, it often leads to false negatives, and there may be few labeled association p-values in the whole genome that can meet this standard, hence, we used the FDR method. Finally, a total of six SNPs significantly related to LMA were identified in this study (Table 2). Moreover, the Manhattan plot of the GWAS result showed the significance of the association between 35,426 SNPs and LMA (Figure 1a). In the Manhattan plots, the -log10 p-values of the quantified SNPs were plotted against their genomic positions. WU_10.2_8_104760632 located on Sus scrofa chromosome (SSC) 8, explained 0.65% of the phenotypic variance of LMA. MARC 0094338, located on SSC 12, accounted for 1.30% of the phenotypic variance of LMA, and ASGA0072817, located on SSC 16, accounted for 2.8% of the phenotypic variance of LMA. The other three SNPs (WU_10.2_14_144250775, ALGA0090878, and ALGA0095241) are located respectively on SSC14, SSC16, and SSC17 (Table 2). We picked the second most significant SNP (MARC0094338) as the top SNP, because there was no previous research on the most significant locus (WU_10.2_8_104760632) and it was not found in the NCBI database. For the SNP MARC0094338, 101 individuals with the AA genotype had higher LMA phenotype values than 278 pigs with the AG genotype and 132 with the GG genotype (Figure 1c). The frequencies of the three genotypes of MARC0094338 (AA, AG, and GG) were 19.8%, 54.4%, and 25.8%, and the mean values of LMA were 74.57, 73.20, and 71.93 cm2. In summary, according to genotype counts and trends of MARC0094338s, the dominant allele frequency of this locus was not high in the current analysis, but it had a positive effect on LMA. This study also indicated that this locus could be continuously selected in the future to improve LMA; besides, the Q-Q plot did not show any sign of inflation (Figure 1b).

Candidate gene search and functional analysis

According to the Sus scrofa 11.1 genome assembly, 11 functional genes were targeted (Table 2). Then, these genes were used to conduct KEGG pathways and GO analyses. In brief, the most significant pathway for LMA was related to the synthesis and degradation of ketone bodies (Supplementary Figure S1a). QTL enrichment analysis showed that meat, carcass, health, and exterior traits were mainly harbored in this pathway (Supplementary Figure S1b). The top GO term of LMA annotated genes were related to the regulation of histone acetylation and growth hormone receptor (GHR) signaling pathway. Given that numerous genes are involved in important pathways and biological processes, functional annotations mentioned in the NCBI database, and a number of earlier published studies were examined and verified; hence, two genes, including 3-oxoacid Co A-transferase 1 (OXCT1) and GHR, with biological functions such as increasing available amino acids in skeletal muscle, fat deposition, were selected as promising candidates for LMA [20].

DISCUSSION

Proper assessment of pig-related indicators is the basis for determining the level of pig performance and growth-related traits. LMA is an influential trait of commercial pigs, affecting production performance, regarded as carcass traits, and plays essential roles in the determination of growth traits [33]. Compared to previous studies, we used cross-bred pigs, which have more complex genome than that of a purebred pig; these were subjected to strong positive selection, and the linkage disequilibrium of DLY crossbred was lower [34], and the genomic information obtained using different quality and density of sequencing chips also varied. In addition, various number of individuals and data filtering conditions also lead to differences in GWAS analyses. In pigs, the major genes specific to the LMA have not yet been identified, and our results are an addition to this area. For pig breeding in the future, it is necessary to clarify the genetic mechanism of LMA. Herein, we performed single-locus GWAS using 511 DLY individuals and identified six loci significantly associated with LMA. In the actual breeding for LMA selection, the SNPs results which we identified through GWAS can be taken as prior biological information into genomic models such as the genomic feature BLUP approach (GFBLUP) [35]. Moreover, recently, researchers proposed several new procedures for calculating SNP weights in wssGBLUP (an extension of ssGBLUP) that can be effective in improving both the accuracy of genomic estimated breeding values and SNP effects [36]. As far as we know, GWAS studies on LMA using DLY populations are still in the minority [37], and actual production could be better served using DLY for the GWAS of LMA. Understanding the genetic basis of phenotypic variation in DLY pigs will help to improve the effectiveness of purebred selection [34]. Other significant SNPs in chromosomes were found for the first time in the present study. This study contributes both to molecular breeding for DLY carcass traits and improves our knowledge of the loci or genes for LMA in commercial pigs.
The GHR gene located at the ASGA0072817 locus of SSC 16 is found to have different degrees of influence on the expression of GHR in the longissimus dorsi and trapezius muscles due to moderate dietary restriction in adults [37], indicating that the GHR gene affects the growth of the longissimus dorsi muscle. The GHR is a single-chain, 620 amino acids transmembrane protein [37]. Hematopoietic, a member of the hematopoietic factor (Cytokine) receptor superfamily, is synthesized and secreted in animals by eosinophils in the anterior pituitary gland and plays a very crucial role in animal growth, development, reproduction, lactation, and immunity [38]. GHR studies in pigs revealed that the abundance of GHR mRNA increased in the longissimus dorsi during mild postpartum malnutrition. Litters were weaned at week 3, and muscle specific GHR expression was assessed for the next three weeks based on optimal or low food intake, resulting in the expression of the GHR gene and its regulation in mild malnutrition. GHR is related to metabolism, contraction, and specific functions of different muscles [39].
Another locus worthy of attention was ASGA0072817, located on SSC 16. The gene OXCT1, also known as the succinyl-CoA:3-ketoacid CoA transferase (SCOT) gene, located near this locus, encodes a key enzyme in ketone metabolism in the body and is also a catalytic switch for ketone decomposition and speed limit steps. In previous studies, such as in Chinese Holstein cows, OXCT1 was identified as a new candidate gene that may affect milk fatty acid synthesis [40,41]. In the lactation period of the yak, the OXCT1 gene was observed to be significantly up-regulated in the mammary gland tissue [41]. The OXCT1 gene can also have a role in adipose tissue and adipocytes. In the adipose tissue of people with higher fat content, regardless of whether its metabolic function is normal or not, the expression of the OXCT1 gene is lower than in people with lower body fat content [42]. In pigs, long non-coding RNAs are involved in important cellular activities in pigs, such as epigenetics, engraftment, and cell growth. Transcriptome analysis was performed in some studies on the back adipose tissue of Korean native pig×Yorkshire pig; KEGG analysis showed that OXCT1 was significantly enriched and involved in the formation of fat [43].
Interestingly, for other genes near significant locus, although some studies have reported that they influence LMA or adipose muscle in pigs, others have reported that these genes are related to cancer or tumor diseases in human pathology. The myocardin (MYOCD) gene locus near the MARC0094338 on SSC 12 is associated with lung cancer [44], the ARho GTPase activating protein 44 (ARHGAP44) gene is associated with tumors [45], the ElaC Ribonuclease Z 2 (ELAC2) gene is reported to be associated with prostate cancer [46], the mitogen-activated protein kinase kinase 4 (MAP2K4) gene is associated with breast cancer [47], F-Box protein 4 (FBXO4) located near ASGA0072817 on SSC 16 is associated with cancer [48], and fibrillarin like 1 (FBLL1) located near WU_10.2_14_144250775 on SSC 14 is associated with liver cancer [49]. In addition, the arginyl-TRNA synthetase 1 (RARS1) gene has been shown to be associated with various diseases of the central nervous system [50], slit guidance ligand 3 (SLIT3) located near ALGA0090878 on SSC16 is associated with the bone loss [51], and the receptor activator of NF-Kb 3 (RANK3) gene is associated with alcoholic femoral head necrosis [52]. While these genes are currently poorly studied in pigs, but do not exclude the possibility of their association with LMA, the OXCT1 gene has been well-documented to play a pivotal role in supporting malignancy [53], with studies in China pointing to its association with adipose development. The population used in this study was DLY ternary crossbred pigs, which has certain limitations in comparison with the reference genome [20]; thus, these genes have great potential to be discovered in the future.

CONCLUSION

In this study, 35,426 SNPs from 511 DLY pigs were used for GWAS analysis. Six significant SNPs were associated with LMA, and two candidate genes (GHR and OXCT1) have been found that maybe influence LMA. In addition, other genes (MYOCD, ARHGAP44, ELAC2, MAP2K4, FBXO4, FBLL1, RARS1, SLIT3, and RANK3) are pending in the future. It is hoped that the results of this study can be used to determine the LMA of pigs and can provide a reference for the genetic improvement of traits.

Notes

CONFLICT OF INTEREST

We certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript. Wu Z is an employee of Guangdong Zhongxin Breeding Technology Co., Ltd..

FUNDING

This study was supported by the Guangdong Province Key Area R&D Program (2022B0202090002), the Natural Science Foundation of Guangdong Province (2018B030313011), and the Tip-top Scientific and Technical Innovative Youth Talents of Guangdong Special Support Program (2019BT02N630). The funding agencies had no role in the study design, data collection and analysis, publication decision, or manuscript preparation.

SUPPLEMENTARY MATERIAL

Supplementary file is available from: https://doi.org/10.5713/ab.22.0407
Supplementary Figure S1. Kyoto encyclopedia of genes and genomes (KEGG) and gene ontology (GO) analysis.
ab-22-0407-Supplementary-Fig-1.pdf
Supplementary Table S1. p-values for fixed affects
ab-22-0407-Supplementary-Table-1.pdf

Figure 1
Manhattan, Quantile–quantile plots and boxplot. (a) Manhattan plot of genome-wide association studies (GWAS) result for loin muscle area (LMA). (b) Quantile–quantile (Q–Q) plot of genome-wide association study for LMA in the Duroc×(Landrace×Yorkshire) crossbred pigs (DLY) population. (c) Boxplot of genotypes of LMA. This boxplot presents the genotype frequencies for MARC0094338, and the counts of three genotypes (AA, AG, and GG) were 101, 278, and 132.
ab-22-0407f1.jpg
Table 1
Descriptive statistics and heritability for LMA
Traits Number Unit Mean (±SD) Min Max C.V./% h21)
LMA 511 cm2 73.82±9.33 45.96 107.21 12.6 0.35±0.096

LMA, loin muscle area; N, number; SD, standard deviation; Min, minimum; Max, maximum; CV, coefficient of variation; h2, heritability.

1) Heritability (standard error) of LMA values.

Table 2
Description of SNPs significantly associated with LMA
Chromosome SNPID Position (bp) EPV (%) p-value Gene1)
8 WU_10.2_8_104760632 97,865,933 0.6365 1.30E-05 -
12 MARC0094338 56,959,881 1.3021 5.39E-05 ARHGAP44, ELAC2, MAP2K4
16 ASGA0072817 26,426,528 2.8139 5.85E-05 RIMOC1, FBX04,GHR, OXCT1
14 WU_10.2_14_144250775 132,664,263 1.4557 8.99E-05 -
17 ALGA0095241 43,212,985 1.8460 1.11E-04 MAFB, FBLL1, RARS1
16 ALGA0090878 54,945,912 0.9760 1.12E-04 RANK3

SNP, single nucleotide polymorphism; LMA, loin muscle area; EPV, the proportion of the phenotypic variance explained by significant SNP; ARHGAP44, ARho GTPase activating protein 44; ELAC2, ElaC Ribonuclease Z 2; MAP2K4, mitogen-activated protein kinase kinase 4; RIMOC1, RAB7A interacting MON1-CCZ1 complex Subunit 1; FBX04, F-Box protein 4; GHR, growth hormone receptor; OXCT1, 3-oxoacid Co A-transferase 1; MAFB, MAF BZIP transcription factor B; FBLL1, fibrillarin like 1; RARS1, arginyl-TRNA synthetase 1; RANK3, receptor activator of NF-Kb 3.

1) The bold genes in the table are candidate genes.

REFERENCES

1. Wang K, Liu D, Hernandez-Sanchez J, et al. Genome wide association analysis reveals new production trait genes in a male Duroc population. Plos One 2015; 10:e139207https://doi.org/10.1371/journal.pone.0139207
crossref pmid pmc
2. Medeiros DOSR, Bonvino SN, de Oliveira FB, et al. Genome-wide association study for carcass traits in an experimental nelore cattle population. Plos One 2017; 12:e169860https://doi.org/10.1371/journal.pone.0169860
crossref pmid pmc
3. Friesen KG, Nelssen JL, Goodband RD, et al. The effect of dietary lysine on growth, carcass composition, and lipid metabolism in high-lean growth gilts fed from 72 to 136 kilograms. J Anim Sci 1995; 73:3392–401. https://doi.org/10.2527/1995.73113392x
crossref pmid
4. Mclaren DG, Mckeith FM, Novakofski J. Prediction of carcass characteristics at market weight from serial real-time ultrasound measures of backfat and loin eye area in the growing pig. J Anim Sci 1989; 67:1657–67. https://doi.org/10.2527/jas1989.6771657x
crossref
5. Godinho RM, Bergsma R, Silva FF, et al. Genetic correlations between feed efficiency traits, and growth performance and carcass traits in purebred and crossbred pigs. J Anim Sci 2018; 96:817–29. https://doi.org/10.1093/jas/skx011
crossref pmid pmc
6. Kuhlers DL, Nadarajah K, Jungst SB, Anderson BL. Genetic selection for real-time ultrasound loin eye area in a closed line of Landrace pigs. Livest Prod Sci 2001; 72:225–31. https://doi.org/10.1016/S0301-6226(01)00222-6
crossref
7. Suzuki K, Kadowaki H, Shibata T, Uchida H, Nishida A. Selection for daily gain, loin-eye area, backfat thickness and intramuscular fat based on desired gains over seven generations of Duroc pigs. Livest Prod Sci 2005; 97:193–202. https://doi.org/10.1016/j.livprodsci.2005.04.007
crossref
8. Onteru SK, Gorbach DM, Young JM, Garrick DJ, Dekkers JCM, Rothschild MF. Whole genome association studies of residual feed intake and related traits in the pig. Plos One 2013; 8:e61756https://doi.org/10.1371/journal.pone.0061756
crossref pmid pmc
9. Thomsen H, Lee HK, Rothschild MF, Malek M, Dekkers JCM. Characterization of quantitative trait loci for growth and meat quality in a cross between commercial breeds of swine. J Anim Sci 2004; 82:2213–28. https://doi.org/10.2527/2004.8282213x
crossref pmid
10. Hu ZL, Park CA, Reecy JM. Bringing the animal QTLdb and CorrDB into the future: meeting new challenges and providing updated services. Nucleic Acids Res 2022; 50:D956–61. https://doi.org/10.1093/nar/gkab1116
crossref pmid pmc
11. Pearson TA, Manolio TA. How to interpret a genome-wide association study. J Am Med Assoc 2008; 299:1335–44. https://doi.org/10.1001/jama.299.11.1335
crossref
12. Andersson L. Genome-wide association analysis in domestic animals: a powerful approach for genetic dissection of trait loci. Genetica 2009; 136:341–9. https://doi.org/10.1007/s10709-008-9312-4
crossref pmid
13. Ramos AM, Crooijmans RP, Affara NA, et al. Design of a high density SNP genotyping assay in the pig using SNPs identified and characterized by next generation sequencing technology. Plos One 2009; 4:e6524https://doi.org/10.1371/journal.pone.0006524
crossref pmid pmc
14. Fernandez AI, Perez-Montarelo D, Barragan C, et al. Genome-wide linkage analysis of QTL for growth and body composition employing the PorcineSNP60 BeadChip. BMC Genet 2012; 13:41https://doi.org/10.1186/1471-2156-13-41
crossref pmid pmc
15. Fontanesi L, Schiavo G, Galimberti G, et al. A genome wide association study for backfat thickness in Italian Large White pigs highlights new regions affecting fat deposition including neuronal genes. BMC Genomics 2012; 13:583https://doi.org/10.1186/1471-2164-13-583
crossref pmid pmc
16. Sahana G, Kadlecova V, Hornshoj H, Nielsen B, Christensen OF. A genome-wide association scan in pig identifies novel regions associated with feed efficiency trait. J Anim Sci 2013; 91:1041–50. https://doi.org/10.2527/jas.2012-5643
crossref pmid
17. He Y, Ma J, Zhang F, et al. Multi-breed genome-wide association study reveals heterogeneous loci associated with loin eye area in pigs. J Appl Genet 2016; 57:511–8. https://doi.org/10.1007/s13353-016-0351-8
crossref pmid
18. Ding R, Zhuang Z, Qiu Y, et al. A composite strategy of genome-wide association study and copy number variation analysis for carcass traits in a Duroc pig population. BMC Genomics 2022; 23:590https://doi.org/10.1186/s12864-022-08804-1
crossref pmid pmc
19. Fan B, Onteru SK, Du ZQ, Garrick DJ, Stalder KJ, Rothschild MF. Genome-wide association study identifies Loci for body composition and structural soundness traits in pigs. Plos One 2011; 6:e14726https://doi.org/10.1371/journal.pone.0014726
crossref pmid pmc
20. Zhuang Z, Li S, Ding R, et al. Meta-analysis of genome-wide association studies for loin muscle area and loin muscle depth in two Duroc pig populations. Plos One 2019; 14:e218263https://doi.org/10.1371/journal.pone.0218263
crossref pmid pmc
21. Ding R, Yang M, Wang X, et al. Genetic architecture of feeding behavior and feed efficiency in a Duroc pig population. Front Genet 2018; 9:220https://doi.org/10.3389/fgene.2018.00220
crossref pmid pmc
22. Purcell S, Neale B, Todd-Brown K, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 2007; 81:559–75. https://doi.org/10.1086/519795
crossref pmid pmc
23. Zhou X, Stephens M. Genome-wide efficient mixed-model analysis for association studies. Nat Genet 2012; 44:821–4. https://doi.org/10.1038/ng.2310
crossref pmid pmc
24. Matukumalli LK, Lawley CT, Schnabel RD, et al. Development and characterization of a high density SNP genotyping assay for cattle. Plos One 2009; 4:e5350https://doi.org/10.1371/journal.pone.0005350
crossref pmid pmc
25. Li S, Qian J, Yang Y, et al. GWAS identifies novel susceptibility loci on 6p21.32 and 21q21.3 for hepatocellular carcinoma in chronic hepatitis B virus carriers. Plos Genet 2012; 8:e1002791https://doi.org/10.1371/journal.pgen.1002791
crossref pmid pmc
26. Lee JN, Wang Y, Xu YO, Li YC, Tian F, Jiang MF. Characterisation of gene expression related to milk fat synthesis in the mammary tissue of lactating yaks. J Dairy Res 2017; 84:283–8. https://doi.org/10.1017/S0022029917000413
crossref pmid
27. Ito K, Murphy D. Application of ggplot2 to pharmacometric graphics. CPT Pharmacometrics Syst Pharmacol 2013; 2:e79https://doi.org/10.1038/psp.2013.56
crossref pmid pmc
28. Wang Y, Ding X, Tan Z, et al. Genome-wide association study of piglet uniformity and farrowing interval. Front Genet 2017; 8:194https://doi.org/10.3389/fgene.2017.00194
crossref pmid pmc
29. Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. Am J Hum Genet 2011; 88:76–82. https://doi.org/10.1016/j.ajhg.2010.11.011
crossref pmid pmc
30. Bu D, Luo H, Huo P, et al. KOBAS-i: intelligent prioritization and exploratory visualization of biological functions for gene enrichment analysis. Nucleic Acids Res 2021; 49:W317–25. https://doi.org/10.1093/nar/gkab447
crossref pmid pmc
31. Fonseca PAS, Suarez-Vega A, Marras G, Canovas A. GALLO: An R package for genomic annotation and integration of multiple data sources in livestock for positional candidate loci. Gigascience. 2020. 9:giaa149https://doi.org/10.1093/gigascience/giaa149
crossref pmid pmc
32. Xing K, Zhu F, Zhai L, et al. Identification of genes for controlling swine adipose deposition by integrating transcriptome, whole-genome resequencing, and quantitative trait loci data. Sci Rep 2016; 6:23219https://doi.org/10.1038/srep23219
crossref pmid pmc
33. Li J, Peng S, Zhong L, et al. Identification and validation of a regulatory mutation upstream of the BMP2 gene associated with carcass length in pigs. Genet Sel Evol 2021; 53:94https://doi.org/10.1186/s12711-021-00689-0
crossref pmid pmc
34. Sarup P, Jensen J, Ostersen T, Henryon M, Sorensen P. Increased prediction accuracy using a genomic feature model including prior information on quantitative trait locus regions in purebred Danish Duroc pigs. BMC Genet 2016; 17:11https://doi.org/10.1186/s12863-015-0322-9
crossref pmid pmc
35. Zhang X, Lourenco D, Aguilar I, Legarra A, Misztal I. Weighting strategies for single-step genomic BLUP: an iterative approach for accurate calculation of GEBV and GWAS. Front Genet 2016; 7:151https://doi.org/10.3389/fgene.2016.00151
crossref pmid pmc
36. Zhang H, Zhuang Z, Yang M, et al. Genome-wide detection of genetic loci and candidate genes for body conformation traits in Duroc x Landrace x Yorkshire crossbred pigs. Front Genet 2021; 12:664343https://doi.org/10.3389/fgene.2021.664343
crossref pmid pmc
37. Combes S, Louveau I, Bonneau M. Moderate food restriction affects skeletal muscle and liver growth hormone receptors differently in pigs. J Nutr 1997; 127:1944–9. https://doi.org/10.1093/jn/127.10.1944
crossref pmid
38. Dettori ML, Pazzola M, Paschino P, Amills M, Vacca GM. Association between the GHR, GHRHR, and IGF1 gene polymorphisms and milk yield and quality traits in Sarda sheep. J Dairy Sci 2018; 101:9978–86. https://doi.org/10.3168/jds.2018-14914
crossref pmid
39. Katsumata M, Cattaneo D, White P, Burton KA, Dauncey MJ. Growth hormone receptor gene expression in porcine skeletal and cardiac muscles is selectively regulated by postnatal undernutrition. J Nutr 2000; 130:2482–8. https://doi.org/10.1093/jn/130.10.2482
crossref pmid
40. Fukao T, Lopaschuk GD, Mitchell GA. Pathways and control of ketone body metabolism: on the fringe of lipid biochemistry. Prostaglandins Leukot Essent Fatty Acids 2004; 70:243–51. https://doi.org/10.1016/j.plefa.2003.11.001
crossref pmid
41. Li C, Sun D, Zhang S, et al. Genome wide association study identifies 20 novel promising genes associated with milk fatty acid traits in Chinese Holstein. Plos One 2014; 9:e96186https://doi.org/10.1371/journal.pone.0096186
crossref pmid pmc
42. Badoud F, Lam KP, Dibattista A, et al. Serum and adipose tissue amino acid homeostasis in the metabolically healthy obese. J Proteome Res 2014; 13:3455–66. https://doi.org/10.1021/pr500416v
crossref pmid
43. Kumar H, Srikanth K, Park W, et al. Transcriptome analysis to identify long non coding RNA (lncRNA) and characterize their functional role in back fat tissue of pig. Gene 2019; 703:71–82. https://doi.org/10.1016/j.gene.2019.04.014
crossref pmid
44. Zhou Q, Chen W, Fan Z, et al. Targeting hyperactive TGFBR2 for treating MYOCD deficient lung cancer. Theranostics 2021; 11:6592–606. https://doi.org/10.7150/thno.59816
crossref pmid pmc
45. Xu J, Jiao J, Xu W, et al. Mutant p53 promotes cell spreading and migration via ARHGAP44. Sci China Life Sci 2017; 60:1019–29. https://doi.org/10.1007/s11427-016-9040-8
crossref pmid
46. Schroeder C, Navid-Hill E, Meiners J, et al. Nuclear ELAC2 overexpression is associated with increased hazard for relapse after radical prostatectomy. Oncotarget 2019; 10:4973–86. https://doi.org/10.18632/oncotarget.27132
crossref pmid pmc
47. Liu S, Huang J, Zhang Y, Liu Y, Zuo S, Li R. MAP2K4 interacts with Vimentin to activate the PI3K/AKT pathway and promotes breast cancer pathogenesis. Aging (Albany NY) 2019; 11:10697–710. https://doi.org/10.18632/aging.102485
crossref pmid pmc
48. Qie S. The E3 ubiquitin ligase Fbxo4 functions as a tumor suppressor: its biological importance and therapeutic perspectives. Cancers (Basel) 2022; 14:2133https://doi.org/10.3390/cancers14092133
crossref pmid pmc
49. Wang L, Zhou N, Qu J, Jiang M, Zhang X. Identification of an RNA binding protein-related gene signature in hepatocellular carcinoma patients. Mol Med 2020; 26:125https://doi.org/10.1186/s10020-020-00252-5
crossref pmid pmc
50. Li G, Eriani G, Wang ED, Zhou XL. Distinct pathogenic mechanisms of various RARS1 mutations in Pelizaeus-Merzbacher-like disease. Sci China Life Sci 2021; 64:1645–60. https://doi.org/10.1007/s11427-020-1838-2
crossref pmid
51. Xu R, Yallowitz A, Qin A, et al. Targeting skeletal endothelium to ameliorate bone loss. Nat Med 2018; 24:823–33. https://doi.org/10.1038/s41591-018-0020-z
crossref pmid pmc
52. Wang T, Wang F, Liu T, et al. OPG/RANKL/RANK gene methylation among alcohol-induced femoral head necrosis in northern Chinese men. J Orthop Surg Res 2021; 16:223https://doi.org/10.1186/s13018-021-02356-y
crossref pmid pmc
53. Zhang S, Xie C. The role of OXCT1 in the pathogenesis of cancer as a rate-limiting enzyme of ketone body metabolism. Life Sci 2017; 183:110–5. https://doi.org/10.1016/j.lfs.2017.07.003
crossref pmid


Editorial Office
Asian-Australasian Association of Animal Production Societies(AAAP)
Room 708 Sammo Sporex, 23, Sillim-ro 59-gil, Gwanak-gu, Seoul 08776, Korea   
TEL : +82-2-888-6558    FAX : +82-2-888-6559   
E-mail : editor@animbiosci.org               

Copyright © 2024 by Asian-Australasian Association of Animal Production Societies.

Developed in M2PI

Close layer
prev next