Next Article in Journal
Challenges in the Application of African Swine Fever Vaccines in Asia
Previous Article in Journal
The Influence of High-Concentrate Diet Supplemented with Tannin on Growth Performance, Rumen Fermentation, and Antioxidant Ability of Fattening Lambs
Previous Article in Special Issue
Phenotypic and Genetic Analyses of Mastitis, Endometritis, and Ketosis on Milk Production and Reproduction Traits in Chinese Holstein Cattle
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genome-Wide Association Study of Conformation Traits in Brazilian Holstein Cattle

by
Emanueli F. P. Silva
1,
Rita C. Gaia
1,
Henrique A. Mulim
2,
Luís Fernando Batista Pinto
3,
Laiza H. S. Iung
4,
Luiz F. Brito
2 and
Victor B. Pedrosa
1,2,5,*
1
Department of Animal Sciences, State University of Ponta Grossa, Ponta Grossa 84010-330, PR, Brazil
2
Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
3
Department of Animal Sciences, Federal University of Bahia, Salvador 40026-010, BA, Brazil
4
Neogen Corporation, Pindamonhangaba 12412-800, SP, Brazil
5
Neogen Corporation, Biotechnology Research, Lincoln, NE 68504, USA
*
Author to whom correspondence should be addressed.
Animals 2024, 14(17), 2472; https://doi.org/10.3390/ani14172472 (registering DOI)
Submission received: 18 July 2024 / Revised: 12 August 2024 / Accepted: 20 August 2024 / Published: 25 August 2024
(This article belongs to the Collection Advances in Cattle Breeding, Genetics and Genomics)

Abstract

:

Simple Summary

Conformation traits have been a key breeding goal in dairy cattle for many years due to their relationship with animal productivity and other relevant characteristics. Therefore, the primary objectives of this study were to conduct genome-wide association studies (GWASs) for conformation traits and identify candidate genes and metabolic pathways associated with these traits in Brazilian Holstein cattle. Phenotypic, pedigree, and genomic (100 K SNP chip) data from 2339 Holstein animals raised across multiple Brazilian states were used for this study. Thirty-six significant SNPs were identified for five composite traits. These genomic markers overlap with candidate genes influencing biological processes such as myogenesis, adipogenesis, and angiogenesis. These findings contribute to unraveling the genetic background of conformation traits in Brazilian Holstein cattle.

Abstract

The linear conformation of animals exerts an influence on health, reproduction, production, and welfare, in addition to longevity, which directly affects the profitability of milk-producing farms. The objectives of this study were (1) to perform genome-wide association studies (GWASs) of conformation traits, namely the Rump, Feet and Legs, Mammary System, Dairy Strength, and Final Classification traits, and (2) to identify genes and related pathways involved in physiological processes associated with conformation traits in Brazilian Holstein cattle. Phenotypic and genotypic data from 2339 Holstein animals distributed across the states of Rio Grande do Sul, Paraná, São Paulo, and Minas Gerais were used. The genotypic data were obtained with a 100 K SNP marker panel. The single-step genome-wide association study (ssGWAS) method was employed in the analyses. Genes close to a significant SNP were identified in an interval of 100 kb up- and downstream using the Ensembl database available in the BioMart tool. The DAVID database was used to identify the main metabolic pathways and the STRING program was employed to create the gene regulatory network. In total, 36 significant SNPs were found on 15 chromosomes; 27 of these SNPs were linked to genes that may influence the traits studied. Fourteen genes most closely related to the studied traits were identified, as well as four genes that showed interactions in important metabolic pathways such as myogenesis, adipogenesis, and angiogenesis. Among the total genes, four were associated with myogenesis (TMOD2, TMOD3, CCND2, and CTBP2), three with angiogenesis (FGF23, FGF1, and SCG3), and four with adipogenesis and body size and development (C5H12orf4, CCND2, EMILIN1, and FGF6). These results contribute to a better understanding of the biological mechanisms underlying phenotypic variability in conformation traits in Brazilian Holstein cattle.

1. Introduction

Genetic selection has played a major role in increasing the sustainability of the dairy farming sector around the world. Production efficiency has improved over the last decade, with an expressive increase in milk production over the past few decades [1]. However, concern about exclusively selecting animals with a genetic profile aimed at increasing milk production has led to unbalanced genetic progress between traits of economic interest [2]. Among these traits, linear conformation traits show a positive genetic correlation with milk production traits in dairy cattle [2,3]. Furthermore, they influence important aspects of dairy farming, including animal health, udder health, and reproductive traits such as calving ease. Thus, conformation traits are associated with both productive and functional traits [4].
Conformation traits have a correlation (0.15 to 0.40) with cows’ functional longevity [5]. Significant single-nucleotide polymorphisms (SNPs) and candidate genes linked to udder quality, body conformation, and fitness in different Holstein cattle populations have been found by genome-wide association studies (GWASs) for conformation traits. SNPs related to udder structure features, such as anterior udder attachment, udder depth, and posterior udder attachment height, have been reported in studies evaluating Chinese Holstein cattle [6,7]. Furthermore, SNPs associated with body depth, fore udder attachment, and overall score have been identified in Czech Holsteins [8]. The adaptation of animals to different environments, diseases, and parasites allows the identification of genes involved in the adaptive process within the species’ genome [9]. Therefore, the evaluation of breeds in different environments is important. Some studies using the GWAS approach in Brazilian Holstein and Girolando dairy cattle breeds [10,11,12] have reported important genomic regions associated with milk production traits, but such results have not yet been reported for conformation traits.
In Brazilian Holstein cattle, there are some important scores used in morphological evaluations. The “Final Classification” (FC) score assess the animal’s general body structure [13]. The “Dairy Strength” (DS) trait assesses the stature, chest width, body depth, angularity, body condition, and topline leveling of the animal, enabling an evaluation of its body balance and production capacity [14], as well-conformed cows likely have greater productive potential and functional longevity. The “Rump” trait is based on a group of variables including rump width, rump angle, loin strength, udder width, and udder attachment [14]. “Feet and Legs” (FL) is assessed based on bone quality, side- and rear-view observations of the legs, heel depth, and hoof angle, thus assessing locomotion characteristics. The “Mammary System” (MS) score includes udder height, udder width, teat placement observed from the rear view, central ligament, udder texture, udder depth, udder attachment, front teat placement, and teat length. MS is associated with cow longevity, health, and well-being, and, therefore, is directly linked to economic returns [15].
In artificial selection, the weight attributed to each trait in breeding schemes reflects the herd’s requirements and breeding objectives, as well as consumer preferences and social needs such as aspects involving the environment [5]. The incorporation of genomic data into genomic prediction analyses has resulted in greater genetic progress compared to pedigree-based selection [16]. An important genomic approach is the genome-wide association study (GWAS) method. One of the strategies of this approach is the investigation of genomic variants that are associated with traits of economic interest [7,17] through the exploration of genomic architecture [16] using SNPs as markers. This technique shows good performance in the assessment of the genetic bases of complex traits, i.e., traits influenced by various genes, such as conformation traits [18].
Gene ontology (GO) enrichment analyses are used to determine the roles of genes that overlap or are in linkage disequilibrium with SNPs associated with enriched pathways that may be related to the traits of interest [19]. Several candidate genes, including MTUS1, PRKN, MGST1, MGST2, STXBP6, and many more, have been linked to udder characteristics [6]. Moreover, conformational features of the legs and feet of Holstein cattle have been found to be associated with genes such as ADIPOR2, INPP4A, DNMT3A, and ALDH1A2 [20]. The genetic architecture of conformation traits in Holstein cattle is better understood thanks to these findings, which also enhances animal longevity, productivity, and health in the dairy sector. The objectives of the present study were (1) to perform GWASs of conformation traits, namely the Rump, Feet and Legs, Mammary System, Dairy Strength, and Final Classification traits, and (2) to identify candidate genes involved in biological processes associated with conformation traits in Brazilian Holstein cattle.

2. Materials and Methods

2.1. Phenotypes and Genotypes

We used data on conformation traits measured in 2339 genotyped Holstein cows from herds in the Brazilian states of Minas Gerais, Paraná, Rio Grande do Sul, and São Paulo. Phenotypic data were collected from primiparous cows in 2021 and 2022. Genotypic data were obtained with the GGP Bovine 100K SNP chip (Neogen Corporation, Lincoln, NE, USA). The genetic material was extracted from hair sample follicles using the phenol–chloroform extraction protocol [21]. A total of 3936 animals were included in the pedigree file after data editing.
The composite traits investigated [22] were as follows:
  • FC, denoting the final score of all conformation traits;
  • DS, comprising the animal’s stature, topline leveling, chest width, body depth, angularity, body condition score, bone quality, udder texture, and lumbar strength;
  • Rump, considering the croup angle, croup width, loin strength, and bone quality;
  • FL, considering the legs from a side view, hoof angle, heel depth, and the legs from the back view;
  • MS, comprising the udder depth, udder texture, median ligament, anterior insertion of the udder, anterior teat placement, udder height, udder width, udder placement, and teat length.

2.2. Quality Control

During the genomic quality control process, we removed SNPs with a call rate < 0.90, SNPs located on non-autosomal chromosomes, SNPs with an unknown or duplicated position, SNPs with a minor allele frequency (MAF) < 0.05, and SNPs with high linkage disequilibrium with other SNPs (r2 > 0.995). After quality control, 82,897 markers remained for subsequent GWASs.
Phenotypic records that exceeded three standard deviations from the mean within the contemporary group (herd, year, and season of birth) were considered to be outliers and removed from further analyses.

2.3. Single-Step GWASs

The single-step GWAS (ssGWAS) method was employed for genomic association analyses using the BLUPF90 family of programs [23]. The RENUMF90 module was used to renumber the phenotypic data and genomic markers. The PREGSF90 module [24] was used for performing genotypic quality control and structuring the relationship matrix, while BLUPF90 [23] was used for the processing of mixed-model equations. Finally, the postGSF90 module was used to back-solve genomic estimated breeding values and to present solutions for the effects of SNPs on each trait.
The traits were analyzed using the following animal model:
y = Xb + Za + e,
where y is the vector of phenotypic observations recorded in primiparous cows; X is the incidence matrix that relates the phenotypes to the fixed effects; b is the vector of fixed effects related to the contemporary group (farm, year, and season of birth) and considering the animal age in months as a linear and quadratic covariate; Z is the incidence matrix that relates the additive genetic effect to the phenotypic records; a is a vector of additive genetic effects; and e is the vector of residual effects.
The variances of a and e are represented as follows:
Var   [ a   e ] = H σ 2 a 0 0 I σ 2 a ,
where σ2a is the direct additive genetic variances for each trait; σ2e is the residual variance; H is the matrix that combines pedigree and genomic information [25]; and I is an identity matrix. The inverse of the matrix H is represented by the following equation:
H 1 = A 1 + 0 0 0 G - 1 - A   22 - 1 ,
where A is the relationship matrix based on the pedigree of all animals; A22 is the relationship matrix based on the pedigree of the genotyped animals; and G is the genomic relationship matrix [26].
The following algorithm was used to solve the SNP effects based on the ssGWAS method [27]:
a = Zu,
where a is the vector of breeding values for genotyped individuals calculated using BLUPF90+; Z is a matrix relating individuals to phenotypes; and u is a vector of the effects of SNP markers.
The SNP effects were estimated using the following equation:
û = IZ′(ZIZ′)−1â,
where û is a vector of the effects of SNP markers; I is an identity matrix; Z is a matrix that relates the individuals to the phenotypes. Each SNP was assumed to have an equal variance of allele substitution effects and the SNP effects were assumed to follow the infinitesimal model.
The approximate p-values of each SNP tested were obtained with the POSTGSF90 program implemented in the BLUPF90 package [28], using the following equation [29]:
p i = 2   ( 1 Φ   ( α i S D ( α i ) ) ) ,
where αi is the SNP effect estimate; SD is the standard deviation; and Φ is the standard normal cumulative distribution function. The p-values were generated by back-solving the SNP effects from the estimated breeding values. This approach was possible because fitting the animal as a random effect to generate estimated breeding values is equivalent to fitting all SNPs as random effects and solving those effects directly [30].

2.4. Identification of Genes and Functional Analyses

The genomic regions harboring significant SNPs were explored in order to identify genes associated with the studied traits, considering a p-value significance threshold of <5 × 10−8 [31]. The list of overlapping (or closely related) genes was assembled considering 100 kb up- and downstream of the significant SNP position in the genome, and the previously reported QTLs overlapping with the identified genomic regions were retrieved using the GALLO R package [32]. The Ensembl Genes database and the bovine reference genome ARS-UCD1.2 [33] were used for these functional genomic analyses.
To improve the understanding of the biological processes shared by these annotated genes, enrichment analyses were performed using the functional annotation tools of the DAVID database [34,35]. Additionally, gene networks were generated using the STRING tool [36]. The criteria used to select the genes for analysis were based on the function and products of the gene as described in the literature, and their relationship to the traits under study.

3. Results

3.1. Descriptive Statistics

The descriptive statistics for the Final Classification (FC), Dairy Strength (DS), Rump, Feet and Legs (FL), and Mammary System (MS) traits of Brazilian Holstein cows are presented in Table 1.

3.2. Single-Step GWASs

GWASs of the conformation traits revealed 36 significant SNPs, including 5 for Final Classification, 10 for Dairy Strength, 9 for Rump, 7 for Feet and Legs, and 5 for Mammary System, as shown in Figure 1. The significant SNPs for the five traits were identified on the following chromosomes: FC: BTA10 and BTA26; DS: BTA3, BTA5, BTA7, BTA10, BTA11, BTA12, BTA19, BTA22, and BTA25; Rump: BTA5, BTA10, BTA11, BTA19, and BTA22; FL: BTA2, BTA3, BTA9, BTA14, BTA22, and BTA23; and MS: BTA7, BTA11, BTA12, and BTA28. Twenty-seven of the identified SNPs were linked to known genes and had QTLs related to conformation traits. Table 2 presents the significant SNPs identified for each trait and the genes associated with them.

3.3. Functional Analyses

The GO terms are shown in Table 3. The TMOD2 and TMOD3 genes, which play a role in the formation of striated skeletal muscle [37], were associated with the FC. Sixteen genes (EMILIN1, FGF23, FGF6, TCF23, RAD51AP1, ACTR2, CCND2, MAPRE3, HELB, FRMD4B, TIGAR, C5H12ofr4, KHK, ARL6IP5, LMOD3, and TMAM214) associated with the formation of the Rump component were enriched for the regulation of cellular processes, the regulation of gene expression, signaling pathways, cell differentiation, organ morphogenesis, and other cellular functions.
Seven of these sixteen genes (C5H12orf4, CCND2, EMILIN1, FGF23, FGF6, HELB, and TIGAR) were found to be closely related to the Rump component. For FL, eight genes with functions in the membrane and cytosol were found to be enriched: ABCC10, ADGRL2, OSBPL10, TJAP1, POLH, HIVEP1, XPO5, and ZNF318. Three of these genes (POLH, HIVP1, and ADGRL2) were associated with FL. Two genes were identified for MS, including COL4A2 and FGF1, which have known functions in the extracellular matrix. No enriched genes associated with GO terms were found for DS.

3.4. Gene Interaction Network

Figure 2 illustrates the gene interaction network. Several genes with different interaction intensities were identified for the conformation traits. As shown in Figure 2, there was an interaction between the genes for Rump (FGF6 and FGF23) and MS (FGF1), as well as with the FLT1 gene involved in angiogenesis. This is important since the heart is one of the main components of the cow’s milk production system due to its function as a blood pump [38].
The TMOD2 and TMOD3 genes, related to Final Classification, interact with the SCG3 gene (involved in angiogenesis). The genes most closely related to Rump (CCND2) and FL (HIVP1) show interactions with each other and with the CTBP2 gene, which is a gene involved in the process of myogenesis. In addition to these interactions, the EMILIN and KHK genes are associated with Rump and seem to interact among themselves.

4. Discussion

4.1. Final Classification

The TMOD2 gene (i.e., tropomodulin 2) is located on BTA10 in cattle. This protein is part of the tropomodulin family, which consists of four isoforms: TMOD1, TMOD2, TMOD3, and TMOD4 [39]. The TMOD2 isoform is expressed in neural structures [40,41,42,43] and encodes a neural tropomodulin that regulates the elongation and depolymerization of actin by interacting with tropomyosin [44]. This protein has been suggested to be associated with the formation of new synaptic structures and the extension of neurites (axon or dendrite) [45]. This process is crucial as the formation or extension of axons directly influences their length, connectivity, and overall functionality [46,47].
The TMOD3 gene (i.e., tropomodulin 3), also located on BTA10, is an isoform of the tropomodulin family [39] and is mainly expressed in skeletal muscle tissue. TMOD3 encodes a protein that binds to the pointed ends of thin actin filaments [48], covering and linking these filaments [49]. TMOD3 controls actin filament polymerization [50] and the interaction of actin with tropomyosin [51]. These functions are important for the maintenance of the structure and function of muscle fibers. A recent study has demonstrated associations of this protein with other traits, such as the adaptive response of mice exposed to different atmospheric pressures at different altitudes [49]. Another study associated the TMOD3 gene with growth traits in dromedaries as a result of its involvement in erythrocyte development, pointed-end actin filament capping, and tropomyosin-binding pathways [52].
The participation of the TMOD2 and TMOD3 genes in the process of myogenesis suggests their relationship with muscle development, which can indirectly influence the conformation traits of dairy cattle.
The CTBP2 gene (i.e., C-terminal-binding protein 2), located on BTA26, encodes C-terminal-binding protein 2, a protein involved in various cellular processes such as transcriptional regulation and DNA damage response. The main function exerted by members of the CTBP family is that of a transcriptional corepressor, interacting with transcription factors and affecting DNA transcription. Additionally, this gene acts in organelles such as the Golgi complex and is involved in the differentiation of brown adipose tissue [53] and cell apoptosis [54]. The CTBP2 gene is also involved in the transcriptional regulation of Notch mediators and other signaling pathways [55], thus participating in the control of various biological processes [56]. Given its involvement in cellular processes, the CTBP2 gene affects developmental processes such as myogenesis [57]. Furthermore, some studies suggest that it exerts an influence on the immune [58,59] and reproductive [60,61,62] systems. CTBP2 has also been reported as a candidate gene for milk protein percentage [63] and angularity in Holstein cows [64].
The ABHD1 gene (i.e., abhydrolase domain-containing 1) is located on BTA11. This gene has been found close to an SNP associated with growth traits in Charolais beef cattle [65]. In Criollo dairy cows, this gene was identified in a run of homozygosity (ROH) [66].
The mRNAs of this gene have been detected in diverse cell types, such as smooth-muscle cells, fibroblasts, some types of blood cells, endothelial cells, and epithelial cells, [67] as well as cells of the liver, an organ with the activity of the ABHD1 gene [68]. The liver interferes with body growth and development since it assists and stimulates the growth of cells and tissues such as bones, muscles, and other organs. The proteins/enzymes encoded by this gene are involved in the metabolic processes of fatty-acid degradation in the liver [69,70]. Furthermore, one study on mice revealed that alpha–beta hydrolase-containing domain 1 plays a role in the regulation of plasma levels of lysophosphatidylcholines [71], an important phospholipid component of cell membranes.

4.2. Rump

The function of the C5H12orf4 gene (i.e., chromosome 5 C12orf4), located on BTA5, has not yet been fully elucidated, but it is known to play role in cell differentiation and growth. Studies on cattle have found C5H12orf4 to be a candidate gene for body size [72] and body weight [73] in different breeds. This fact points to a possible relationship of this gene with the body structure of cattle, indicating a direct relationship with linear conformation in dairy cattle.
The CCND2 gene (i.e., cyclin D2), located on BTA5, encodes cyclin D2, a regulatory protein that is part of the cyclin D2/cyclin-dependent kinase 4 (CDK4) complex. This complex is involved in the transition from the G1 phase to the S phase [74,75,76]. Studies indicate participation of this gene in reproduction and in the regulation of reproductive hormones in different species [77,78,79,80,81,82,83,84]. The CCND2 gene has also been associated with the proliferation of pre-adipocytes [85,86] and muscle cells [87]. In cattle, this gene has been identified as a possible influencer of stature and body size [72,88,89] and body conformation [90], as well as of daily weight gain in Holstein cattle [73]. As seen in studies evaluating this gene, CCND2 might be associated with the morphological traits of animals since it is involved in myogenesis and adipogenesis, influencing the composition of body structure.
The EMILIN1 gene (i.e., elastin microfibril interfacer 1), located on BTA11, encodes an extracellular-matrix glycoprotein. EMILIN1 is involved in elastogenesis in different tissues, in addition to its interaction with transforming growth factor beta (TGFβ) [91], a growth factor that plays a role in embryonic development, cell differentiation, hormone secretion, and immune function. This protein is part of the microfibrillar structure of elastic fibers [92], contributing to the elasticity of skin and tissues [93]. Additionally, the EMILIN1 gene has been associated with growth traits in Charolais cattle [65] and with placentation and trophoblast migration and invasion [66,94,95]. A higher expression of this gene is observed in tissues that require greater elasticity, such as the skin and organs like the uterus and mammary glands. This gene can affect the conformation of animals since it has been associated with the growth of cattle. The EMILIN1 gene can also be associated with the maintenance of the Mammary System, which affects dairy cattle.
The FGF23 gene (i.e., fibroblast growth factor 23), located on BTA5, is a member of the fibroblast growth factor (FGF) family. Studies have linked this hormone to different biological processes, such as angiogenesis, morphogenesis, tissue regulation, and oncogenesis [96]; to the cardiovascular system [97]; and to the formation of osteoclasts [98]. Produced mainly in bones, FGF23 is involved in phosphate homeostasis (PO43−) and vitamin D metabolism [99,100,101,102]. PO43− actively participates in various biological processes such as skeletal development, bone mineralization, membrane composition, nucleotide structuring, the maintenance of plasma pH, and cell signaling.
The FGF6 gene (i.e., fibroblast growth factor 6), located on BTA5, is also a member of the FGF family that is involved in different cellular processes. Studies on different cattle breeds have demonstrated the influence of this gene on body size [72] and body conformation [90], as well as suggesting that this gene has a possible effect on muscle mass [103]. Furthermore, it has been suggested that the levels of expression of this gene in adipose tissue are increased in colder environments for better thermogenesis [104]. This gene is involved in biological processes such as angiogenesis, morphogenesis, tissue regulation, and oncogenesis [96], as well as in embryonic development [105]. Studies on fish have reported the influence of FGF6 on muscle tissue hyperplasia [106]. Additionally, a very important role of this protein in myogenesis has been observed in both adult and newborn rats [107]. FGF6 is also involved in muscle regeneration [108,109].
The HELB gene (i.e., DNA helicase B) is located on BTA5 and is expressed during cell replication [110] since it is involved in DNA repair and duplication. As a helicase, it unwinds the double-helix DNA and is therefore mainly found in the nucleus during the G1 phase of the cell cycle [111]. Furthermore, in cattle, this gene plays a role in the adaptation of animals to regions with a tropical climate [112] and to environments characterized by constant high temperatures and high levels of UV intensity [113]. Several SNPs in this gene have been associated with yearling weight in Brahman cattle [9].
The TIGAR gene (i.e., TP53-induced glycolysis-regulator phosphatase) is located on BTA5 and exerts important functions at the cellular level, being involved in glucose metabolism [114,115,116], the regulation of the cell cycle [117], and cellular responses to metabolic stress [118]. This gene is expressed in almost all tissues of the body, with higher levels of expression in muscles, the brain, and the heart [119], and is mainly regulated by the stress-induced transcription factor p53, which is responsible for controlling cellular mechanisms such as the cell cycle and apoptosis [120]. The TIGAR gene encodes a glucose-regulating enzyme that contributes to the formation of nicotinamide adenine dinucleotide phosphate (NADPH) [121]. Furthermore, studies on cattle have associated this gene with body size, body weight, and body structure [122].
The SCG3 gene (i.e., secretogranin III) is located on BTA10 and encodes secretogranin III, a protein that is mainly expressed in the neuroendocrine system and that plays a role in the storage and transport of neurotransmitters [123]. A recent study has indicated involvement of this gene in angiogenesis [124]. SCG3 has also been associated with obesity in humans [125] and puberty in cattle [126,127,128].

4.3. Feet and Legs

The POLH gene (i.e., DNA polymerase eta), located on BTA23, encodes DNA polymerase eta, an enzyme involved in the cell cycle, DNA replication and repair, and cell morphology [129]. Mutations in this gene affect the accuracy of DNA replication, resulting in additional mutations [130,131]. Previous studies in the literature do not establish a direct linkage between the Feet and Legs trait and the genomic regions identified here. However, polymerases play a crucial role in repairing DNA lesions and preventing a wide range of DNA damage, which is vital for embryonic viability [132]. During mitotic division, polymerases, in conjunction with other proteins, ensure the successful completion of DNA replication, which is vital for several biological organizations as well as for body structure [133].
The HIVEP1 gene (i.e., human immunodeficiency virus type I enhancer-binding protein 1) is located on BTA23. The protein encoded by this gene participates in the regulation of genes involved in the inflammatory response [134,135,136].
The ADGRL2 gene (i.e., adhesion G-protein-coupled receptor L2), located on BTA3, has been associated with weight and growth in pigs [137], as well as with the development of intra- and extracellular regions [138]. In humans, this gene has also been associated with the immune system [139].

4.4. Mammary System

The COL4A2 gene (i.e., collagen type IV alpha 2), located on BTA12, encodes the proα2 (IV) protein, a precursor molecule of type IV collagen responsible for providing structural support to tissues and organs such as the endomysium and perimysium [140]. This type of collagen is the main component of basement membranes [141]. Together with other membrane components, this collagen is responsible for incorporation, regulation, and membrane adhesion [142]. Mutations in this gene can cause diseases in different tissues, including muscle tissue [143].
A study on sheep found an increase in the expression of the COL4A2 gene at the end of lactation, which was associated with basal lamina turnover during involution of the mammary glands [144]. In milk-producing Holstein cows, this gene was found to play an important role in maintaining the morphology and function of the mammary glands [145]. A study evaluating different types of diet in Holstein cows found a difference in gene expression depending on the type of feed ingested, which influenced mammary growth and development [146]. Furthermore, recent studies indicate an influence of the COL4A2 gene on fat metabolism [147,148]. According to the traits already related to these genes, their functions are associated with udder constitution/structure in dairy cattle.
The FGF1 gene (i.e., fibroblast growth factor 1), located on BTA7, is a member of the FGF family. The fibroblast growth factor 1 protein plays a role in the regulation of cell proliferation [149], cell division, cell migration, neurogenesis [150,151], and angiogenesis [152]. The FGF1 gene is involved in reproduction [153,154,155,156,157] in bovines. Furthermore, its protein is important for tissue repair [158] considering that the FGF group of proteins participates in the formation of a type of connective tissue.
The FLT1 gene (i.e., vascular endothelial growth factor receptor 1), located on BTA12 and a member of the kinase family, is involved in angiogenesis [159,160,161,162]. Furthermore, recent studies on Holstein cattle have associated this gene with other traits such as udder depth, central ligament, Dairy Strength, and stillbirth risk [163], as well as with the innate immune response [164]. These findings lead us to consider this gene as a candidate for the formation of the traits studied, especially those related to conformation of the Mammary System.

5. Conclusions

We identified 36 significant SNPs and 18 candidate genes associated with 23 metabolic pathways for the five traits evaluated. Among the highlighted genes, the FGF23 gene has been associated with osteogenesis and angiogenesis. The CCDN2, TMOD2, TMOD3, and CTBP2 genes have been associated with myogenesis. Furthermore, the C5H12orf4, FGF6, EMILIN1, and ABHD1 genes have been associated with body size, growth, and weight gain in cattle. The SCG3 and FLT1 genes have been associated with angiogenesis. The HELB and TIGAR genes are involved in the adaptation of animals to different climates and altitudes. In conclusion, we identified various genomic regions associated with five conformation traits in Brazilian Holstein cattle. These findings are important for understanding the molecular bases of conformation traits in dairy cattle.

Author Contributions

V.B.P. conceptualized the experiment. V.B.P. and L.H.S.I. collected the data. V.B.P., E.F.P.S. and R.C.G. analyzed the data. E.F.P.S., V.B.P., H.A.M., L.F.B., L.F.B.P. and L.H.S.I. prepared the original draft and finished the manuscript. V.B.P. was the project administrator and supervised the experiment. All authors have read and agreed to the published version of the manuscript.

Funding

The authors thank the Conselho Nacional de Desenvolvimento Cientıfico e Tecnológico (CNPq-N◦ 303074/2019-1) and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) for providing financial support for this study.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Schmidtmann, C.; Segelke, D.; Bennewitz, J.; Tetens, J.; Thaller, G. Genetic Analysis of Production Traits and Body Size Measurements and Their Relationships with Metabolic Diseases in German Holstein Cattle. J. Dairy Sci. 2022, 106, 421–438. [Google Scholar] [CrossRef] [PubMed]
  2. Miglior, F.; Fleming, A.; Malchiodi, F.; Brito, L.F.; Martin, P.; Baes, C.F. A 100-Year Review: Identification and Genetic Selection of Economically Important Traits in Dairy Cattle. J. Dairy Sci. 2017, 100, 10251–10271. [Google Scholar] [CrossRef] [PubMed]
  3. Xue, X.; Hu, H.; Zhang, J.; Ma, Y.; Han, L.; Hao, F.; Jiang, Y.; Ma, Y. Estimation of Genetic Parameters for Conformation Traits and Milk Production Traits in Chinese Holsteins. Animals 2023, 13, 100. [Google Scholar] [CrossRef]
  4. Sawa, A.; Bogucki, M.; Krężel-Czopek, S.; Neja, W. Relationship between Conformation Traits and Lifetime Production Efficiency of Cows. ISRN Vet. Sci. 2013, 2013, 124690. [Google Scholar] [CrossRef] [PubMed]
  5. Paakala, E.; Martín-Collado, D.; Mäki-Tanila, A.; Juga, J. Variation in the Actual Preferences for AI Bull Traits among Finnish Dairy Herds. J. Anim. Breed. Genet. 2018, 135, 410–419. [Google Scholar] [CrossRef]
  6. Nazar, M.; Abdalla, I.M.; Chen, Z.; Ullah, N.; Liang, Y.; Chu, S.; Xu, T.; Mao, Y.; Yang, Z.; Lu, X. Genome-Wide Association Study for Udder Conformation Traits in Chinese Holstein Cattle. Animals 2022, 12, 2542. [Google Scholar] [CrossRef] [PubMed]
  7. Yan, Z.; Wang, Z.; Zhang, Q.; Yue, S.; Yin, B.; Jiang, Y.; Shi, K. Identification of Whole-Genome Significant Single Nucleotide Polymorphisms in Candidate Genes Associated with Body Conformation Traits in Chinese Holstein Cattle. Anim. Genet. 2020, 51, 141–146. [Google Scholar] [CrossRef]
  8. Čítek, J.; Brzáková, M.; Bauer, J.; Tichý, L.; Sztankóová, Z.; Vostrý, L.; Steyn, Y. Genome-Wide Association Study for Body Conformation Traits and Fitness in Czech Holsteins. Animals 2022, 12, 3522. [Google Scholar] [CrossRef]
  9. Naval-Sánchez, M.; Porto-Neto, L.R.; Cardoso, D.F.; Hayes, B.J.; Daetwyler, H.D.; Kijas, J.; Reverter, A. Selection Signatures in Tropical Cattle Are Enriched for Promoter and Coding Regions and Reveal Missense Mutations in the Damage Response Gene HELB. Genet. Sel. Evol. 2020, 52, 27. [Google Scholar] [CrossRef]
  10. Iung, L.H.S.; Petrini, J.; Ramírez-Díaz, J.; Salvian, M.; Rovadoscki, G.A.; Pilonetto, F.; Dauria, B.D.; Machado, P.F.; Coutinho, L.L.; Wiggans, G.R.; et al. Genome-Wide Association Study for Milk Production Traits in a Brazilian Holstein Population. J. Dairy Sci. 2019, 102, 5305–5314. [Google Scholar] [CrossRef]
  11. Otto, P.I.; Guimarães, S.E.F.; Calus, M.P.L.; Vandenplas, J.; Machado, M.A.; Panetto, J.C.C.; da Silva, M.V.G.B. Single-Step Genome-Wide Association Studies (GWAS) and Post-GWAS Analyses to Identify Genomic Regions and Candidate Genes for Milk Yield in Brazilian Girolando Cattle. J. Dairy Sci. 2020, 103, 10347–10360. [Google Scholar] [CrossRef] [PubMed]
  12. Carrara, E.R.; Dauria, B.D.; Gervásio, I.C.; Silveira, R.M.F.; Rovadoski, G.A.; Petrini, J.; Salvian, M.; Machado, P.F.; Mourão, G.B. Comparison of Marker Effects and Breeding Values at Two Levels at THI for Milk Yield and Quality Traits in Brazilian Holstein Cows. Genes 2023, 14, 17. [Google Scholar] [CrossRef]
  13. Bohlouli, M.; Alijani, S.; Varposhti, M.R. Genetic Relationships among Linear Type Traits and Milk Production Traits of Holstein Dairy Cattle. Ann. Anim. Sci. 2015, 15, 903–917. [Google Scholar] [CrossRef]
  14. Atkins, G. Using Conformational Anatomy to identify functionality in Dairy cows. In Proceedings of the 24th South Western Ontario Dairy Symposium, Woodstock, ON, Canada, 20 February 2007; pp. 37–48. [Google Scholar]
  15. Devani, K.; Plastow, G.; Orsel, K.; Valente, T.S. Genome-Wide Association Study for Mammary Structure in Canadian Angus Cows. PLoS ONE 2020, 15, e0237818. [Google Scholar] [CrossRef] [PubMed]
  16. Massender, E.; Oliveira, H.R.; Brito, L.F.; Maignel, L.; Jafarikia, M.; Baes, C.F.; Sullivan, B.; Schenkel, F.S. Genome-Wide Association Study for Milk Production and Conformation Traits in Canadian Alpine and Saanen Dairy Goats. J. Dairy Sci. 2023, 106, 1168–1189. [Google Scholar] [CrossRef]
  17. Tizioto, P.C.; Coutinho, L.L.; Oliveira, P.S.N.; Cesar, A.S.M.; Diniz, W.J.S.; Lima, A.O.; Rocha, M.I.; Decker, J.E.; Schnabel, R.D.; Mourão, G.B.; et al. Gene Expression Differences in Longissimus Muscle of Nelore Steers Genetically Divergent for Residual Feed Intake. Sci. Rep. 2016, 6, 39493. [Google Scholar] [CrossRef]
  18. Cantor, R.M.; Lange, K.; Sinsheimer, J.S. Prioritizing GWAS Results: A Review of Statistical Methods and Recommendations for Their Application. Am. J. Hum. Genet. 2010, 86, 6–22. [Google Scholar] [CrossRef]
  19. Gebreyesus, G.; Buitenhuis, A.J.; Poulsen, N.A.; Visker, M.H.P.W.; Zhang, Q.; Van Valenberg, H.J.F.; Sun, D.; Bovenhuis, H. Multi-Population GWAS and Enrichment Analyses Reveal Novel Genomic Regions and Promising Candidate Genes Underlying Bovine Milk Fatty Acid Composition. BMC Genom. 2019, 20, 178. [Google Scholar] [CrossRef]
  20. Abdalla, I.M.; Lu, X.; Nazar, M.; Arbab, A.A.I.; Xu, T.; Yousif, M.H.; Mao, Y.; Yang, Z. Genome-Wide Association Study Identifies Candidate Genes Associated with Feet and Leg Conformation Traits in Chinese Holstein Cattle. Animals 2021, 11, 2259. [Google Scholar] [CrossRef]
  21. Sambrook, J.; Fritsch, E.F.; Maniatis, T. Molecular Cloning: A Laboratory Manual, 2nd ed.; Cold Spring Harbor Laboratory Press: Long Island, NY, USA, 1989. [Google Scholar]
  22. Valloto, A.A.; Ribas Neto, P.G. Avaliação da Conformação Ideal das Vacas Leiteiras; SENAR—PR: Curitiba, Brazil, 2012. [Google Scholar]
  23. Misztal, I.; Aguilar, I.; Tsuruta, S.; Masuda, Y.; Lourenco, D.A.L.; Legarra, A. BLUPF90 Suite of Programs for Animal Breeding. In Proceedings of the 11th World Congress of Genetics Applied to Livestock Production, Auckland, New Zealand, 11–16 February 2018; Volume 11, p. 751. [Google Scholar]
  24. Aguilar, I.; Misztal, I.; Tsuruta, S.; Legarra, A.; Wang, H. PREGSF90—POSTGSF90: Computational Tools for the Implementation of Single-Step Genomic Selection and Genome-Wide Association with Ungenotyped Individuals in BLUPF90 Programs. In Proceedings of the 10th World Congress of Genetics Applied to Livestock Production PREGSF90, Vancouver, BC, Canada, 17–22 August 2014. [Google Scholar]
  25. Aguilar, I. Genetic Evaluation Including Phenotypic, Full Pedigree and Genomic Data. Master’s Thesis, University of Georgia, Athens, GA, USA, 2010. [Google Scholar]
  26. Ignacy, M.; Shogo, T.; Daniela, L.; Yutaka, M.; Ignacio, A.; Andres, L.; Zulma, V. Manual for BLUPF90 Family Programs; University of Georgia: Athens, GA, USA, 2018. [Google Scholar]
  27. Wang, H.; Misztal, I.; Aguilar, I.; Legarra, A.; Muir, W.M. Genome-Wide Association Mapping Including Phenotypes from Relatives without Genotypes. Genet. Res. 2012, 94, 73–83. [Google Scholar] [CrossRef]
  28. Masuda, Y. Introduction to BLUPF90 Suite Programs Standard Edition; University of Georgia: Athens, GA, USA, 2019. [Google Scholar]
  29. Aguilar, I.; Legarra, A.; Cardoso, F.; Masuda, Y.; Lourenco, D.; Misztal, I. Frequentist P-Values for Large-Scale-Single Step Genome-Wide Association, with an Application to Birth Weight in American Angus Cattle. Genet. Sel. Evol. 2019, 51, 28. [Google Scholar] [CrossRef] [PubMed]
  30. Garrick, D.J. Equivalent Mixed Model Equations for Genomic Selection. J. Anim. Sci. 2007, 85, 376. [Google Scholar]
  31. Jannot, A.; Ehret, G.; Perneger, T. P < 5 × 10−8 Has Emerged as a Standard of Statistical Significance for Genome-Wide Association Studies. J. Clin. Epidemiol. 2015, 68, 460–465. [Google Scholar] [PubMed]
  32. Fonseca, P.A.S.; Suárez-Vega, A.; Marras, G.; Cánovas, Á. GALLO: An R Package for Genomic Annotation and Integration of Multiple Data Sources in Livestock for Positional Candidate Loci. Gigascience 2020, 9, giaa149. [Google Scholar] [CrossRef]
  33. Lindholm-Perry, A.K.; Meyer, A.M.; Kern-Lunbery, R.J.; Cunningham-Hollinger, H.C.; Funk, T.H.; Keel, B.N. Genes Involved in Feed Efficiency Identified in a Meta-Analysis of Rumen Tissue from Two Populations of Beef Steers. Animals 2022, 12, 1514. [Google Scholar] [CrossRef]
  34. Sherman, B.T.; Hao, M.; Qiu, J.; Jiao, X.; Baseler, M.W.; Lane, H.C.; Imamichi, T.; Chang, W. DAVID: A Web Server for Functional Enrichment Analysis and Functional Annotation of Gene Lists (2021 Update). Nucleic Acids Res. 2022, 50, W216–W221. [Google Scholar] [CrossRef]
  35. Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Systematic and Integrative Analysis of Large Gene Lists Using DAVID. Nat. Protoc. 2009, 4, 44–57. [Google Scholar] [CrossRef]
  36. Szklarczyk, D.; Kirsch, R.; Koutrouli, M.; Nastou, K.; Mehryary, F.; Hachilif, R.; Gable, A.L.; Fang, T.; Doncheva, N.T.; Pyysalo, S.; et al. The STRING Database in 2023: Protein–Protein Association Networks and Functional Enrichment Analyses for Any Sequenced Genome of Interest. Nucleic Acids Res. 2023, 51, D638–D646. [Google Scholar] [CrossRef]
  37. Bouley, J.; Chambon, C.; Picard, B. Mapping of Bovine Skeletal Muscle Proteins Using Two-Dimensional Gel Electrophoresis and Mass Spectrometry. Proteomics 2004, 4, 1811–1824. [Google Scholar] [CrossRef]
  38. Lawrence, R.A. Physiology of Lactation. In Breastfeeding; Elsevier: Amsterdam, The Netherlands, 2022; pp. 58–92. [Google Scholar]
  39. Tolkatchev, D.; Kuruba, B.; Smith, G.E.; Swain, K.D.; Smith, K.A.; Moroz, N.; Williams, T.J.; Kostyukova, A.S. Structural Insights into the Tropomodulin Assembly at the Pointed Ends of Actin Filaments. Protein Sci. 2021, 30, 423–437. [Google Scholar] [CrossRef]
  40. Bao, Y.; Kake, T.; Hanashima, A.; Nomiya, Y.; Kubokawa, K.; Kimura, S. Actin Capping Proteins, CapZ (β-Actinin) and Tropomodulin in Amphioxus Striated Muscle. Gene 2012, 510, 78–86. [Google Scholar] [CrossRef] [PubMed]
  41. Fath, T.; Fischer, R.S.; Dehmelt, L.; Halpain, S.; Fowler, V.M. Tropomodulins Are Negative Regulators of Neurite Outgrowth. Eur. J. Cell Biol. 2011, 90, 291–300. [Google Scholar] [CrossRef] [PubMed]
  42. Moroz, N.; Guillaud, L.; Desai, B.; Kostyukova, A.S. Mutations Changing Tropomodulin Affinity for Tropomyosin Alter Neurite Formation and Extension. PeerJ 2013, 2013, e7. [Google Scholar] [CrossRef] [PubMed]
  43. Yamashiro, S.; Gokhin, D.S.; Kimura, S.; Nowak, R.B.; Fowler, V.M. Tropomodulins: Pointed-End Capping Proteins That Regulate Actin Filament Architecture in Diverse Cell Types. Cytoskeleton 2012, 69, 337–370. [Google Scholar] [CrossRef]
  44. Quintana, A.M.; Yu, H.C.; Brebner, A.; Pupavac, M.; Geiger, E.A.; Watson, A.; Castro, V.L.; Cheung, W.; Chen, S.H.; Watkins, D.; et al. Mutations in THAP11 Cause an Inborn Error of Cobalamin Metabolism and Developmental Abnormalities. Hum. Mol. Genet. 2017, 26, 2838–2849. [Google Scholar] [CrossRef]
  45. Omotade, O.F.; Rui, Y.; Lei, W.; Yu, K.; Hartzell, H.C.; Fowler, V.M.; Zheng, J.Q. Tropomodulin Isoform-Specific Regulation of Dendrite Development and Synapse Formation. J. Neurosci. 2018, 38, 10271–10285. [Google Scholar] [CrossRef]
  46. Taherabadi, S.J.; Rahmati, M. Exercise Training Enhances Expression of Tropomedulin-2 and ADP-Ribosylation Factor 6 in the Cerebellum of Male Wistar Rats. Zahedan J. Res. Med. Sci. 2019, 21, e89902. [Google Scholar] [CrossRef]
  47. Taherabadi, S.J.; Rahmati, M.; Mirnasuri, R.; Kazemi, A. Effect of Exercise Training on Tropomodulin-2 Gene Expression in Cerebellum of Diabetic Rats. Iran. J. Diabetes Obes. 2019, 11, 28–37. [Google Scholar]
  48. Gokhin, D.S.; Tierney, M.T.; Sui, Z.; Sacco, A.; Fowler, V.M. Calpain-Mediated Proteolysis of Tropomodulin Isoforms Leads to Thin Filament Elongation in Dystrophic Skeletal Muscle. Mol. Biol. Cell 2014, 25, 852–865. [Google Scholar] [CrossRef]
  49. Stobdan, T.; Jain, P.P.; Xiong, M.; Bafna, V.; Yuan, J.X.J.; Haddad, G.G. Heterozygous Tropomodulin 3 Mice Have Improved Lung Vascularization after Chronic Hypoxia. Hum. Mol. Genet. 2022, 31, 1130–1140. [Google Scholar] [CrossRef]
  50. Liu, S.; Han, W.; Jiang, S.; Zhao, C.; Wu, C. Integrative Transcriptomics and Proteomics Analysis of Longissimus Dorsi Muscles of Canadian Double-Muscled Large White Pigs. Gene 2016, 577, 14–23. [Google Scholar] [CrossRef] [PubMed]
  51. Nolte, W.; Thaller, G.; Kuehn, C. Selection Signatures in Four German Warmblood Horse Breeds: Tracing Breeding History in the Modern Sport Horse. PLoS ONE 2019, 14, e0215913. [Google Scholar] [CrossRef] [PubMed]
  52. Sani, M.B.; Roudbari, Z.; Karimi, O.; Banabazi, M.H.; Esmaeilkhanian, S.; Asadzadeh, N.; Zare Harofte, J.; Shafei Naderi, A.; Burger, P.A. Gene-Set Enrichment Analysis for Identifying Genes and Biological Activities Associated with Growth Traits in Dromedaries. Animals 2022, 12, 184. [Google Scholar] [CrossRef] [PubMed]
  53. Acosta-Baena, N.; Tejada-Moreno, J.A.; Arcos-Burgos, M.; Villegas-Lanau, C.A. CTBP1 and CTBP2 Mutations Underpinning Neurological Disorders: A Systematic Review. Neurogenetics 2022, 23, 231–240. [Google Scholar] [CrossRef]
  54. Kovi, R.C.; Paliwal, S.; Pande, S.; Grossman, S.R. An ARF/CtBP2 Complex Regulates BH3-Only Gene Expression and P53-Independent Apoptosis. Cell Death Differ. 2010, 17, 513–521. [Google Scholar] [CrossRef]
  55. Mengistie, D.; Suk Kim, K.; Samuel, B. Genome-Wide Signature of Positive Selection in Ethiopian Indigenous and European Beef Cattle Breeds. Res. Sq. 2022, 29, 200151. [Google Scholar] [CrossRef]
  56. Van Hateren, N.; Shenton, T.; Borycki, A.G. Expression of Avian C-Terminal Binding Proteins (Ctbp1 and Ctbp2) during Embryonic Development. Dev. Dyn. 2006, 235, 490–495. [Google Scholar] [CrossRef]
  57. Ponsuksili, S.; Murani, E.; Phatsara, C.; Schwerin, M.; Schellander, K.; Wimmers, K. Porcine Muscle Sensory Attributes Associate with Major Changes in Gene Networks Involving CAPZB, ANKRD1, and CTBP2. Funct. Integr. Genom. 2009, 9, 455–471. [Google Scholar] [CrossRef]
  58. Chen, Z.; Dong, W.H.; Qiu, Z.M.; Li, Q.G. The Monocyte-Derived Exosomal CLMAT3 Activates the CtBP2-P300-NF-ΚB Transcriptional Complex to Induce Proinflammatory Cytokines in ALI. Mol. Ther. Nucleic Acids 2020, 21, 1100–1110. [Google Scholar] [CrossRef]
  59. Wang, J.; Lee, S.; Teh, C.E.Y.; Bunting, K.; Ma, L.; Shannon, M.F. The Transcription Repressor, ZEB1, Cooperates with CtBP2 and HDAC1 to Suppress IL-2 Gene Activation in T Cells. Int. Immunol. 2009, 21, 227–235. [Google Scholar] [CrossRef]
  60. Cochran, S.D.; Cole, J.B.; Null, D.J.; Hansen, P.J. Discovery of Single Nucleotide Polymorphisms in Candidate Genes Associated with Fertility and Production Traits in Holstein Cattle. BMC Genet. 2013, 14, 49. [Google Scholar] [CrossRef] [PubMed]
  61. Liu, A.; Wang, Y.; Sahana, G.; Zhang, Q.; Liu, L.; Lund, M.S.; Su, G. Genome-Wide Association Studies for Female Fertility Traits in Chinese and Nordic Holsteins. Sci. Rep. 2017, 7, 8487. [Google Scholar] [CrossRef] [PubMed]
  62. Valdez-Torres, J.M.; Ahuir, J.A.G.; Castro-Valenzuela, B.E.; Eduviges Burrola-Barraza, M. QTL Analysis Associated to Single Nucleotide Polymorphisms (SNP) Involved in the Dairy Phenotype of Holstein Cattle. Rev. Mex. Cienc. Pecu. 2021, 11, 1192–1207. [Google Scholar] [CrossRef]
  63. Vijayakumar, P.; Bakyaraj, S.; Singaravadivelan, A.; Vasanthakumar, T.; Suresh Vijayakumar, R.P.; Singaravadivelan, A.; Vasanthakumar, T.; Suresh Veterinary, R.; Bakyaraj, S. Meta-Analysis of Mammary RNA Seq Datasets Reveals the Molecular Understanding of Bovine Lactation Biology. Genome 2019, 62, 489–501. [Google Scholar] [CrossRef] [PubMed]
  64. Kolbehdari, D.; Wang, Z.; Grant, J.R.; Murdoch, B.; Prasad, A.; Xiu, Z.; Marques, E.; Stothard, P.; Moore, S.S. A Whole-Genome Scan to Map Quantitative Trait Loci for Conformation and Functional Traits in Canadian Holstein Bulls. J. Dairy Sci. 2008, 91, 2844–2856. [Google Scholar] [CrossRef]
  65. Jahuey-Martínez, F.J.; Parra-Bracamonte, G.M.; Sifuentes-Rincón, A.M.; Martínez-González, J.C.; Gondro; García-Pérez, C.A.; López-Bustamante, L.A. Genomewide Association Analysis of Growth Traits in Charolais Beef Cattle. J. Anim. Sci. 2016, 94, 4570–4582. [Google Scholar] [CrossRef]
  66. Martínez-Rocha, R.; Hidalgo, J.; Cesarani, A.; Ramírez-Valverde, R.; Núñez-Domínguez, R.; García-Muñiz, J.G.; Domínguez-Viveros, J. Assessment of Genetic Diversity, Runs of Homozygosity, and Signatures of Selection in Tropical Milking Criollo Cattle Using Pedigree and Genomic Data. Genes 2022, 13, 1896. [Google Scholar] [CrossRef]
  67. Makalowska, I.; Lin, C.F.; Makalowski, W. Overlapping Genes in Vertebrate Genomes. Comput. Biol. Chem. 2005, 29, 1–12. [Google Scholar] [CrossRef] [PubMed]
  68. Shahzad, K.; Akbar, H.; Vailati-Riboni, M.; Basiricò, L.; Morera, P.; Rodriguez-Zas, S.L.; Nardone, A.; Bernabucci, U.; Loor, J.J. The Effect of Calving in the Summer on the Hepatic Transcriptome of Holstein Cows during the Peripartal Period. J. Dairy Sci. 2015, 98, 5401–5413. [Google Scholar] [CrossRef]
  69. Liu, G.; Wang, L.; Wess, J.; Dean, A. Enhancer Looping Protein LDB1 Regulates Hepatocyte Gene Expression by Cooperating with Liver Transcription Factors. Nucleic Acids Res. 2022, 50, 9195–9211. [Google Scholar] [CrossRef]
  70. Wathes, D.C.; Cheng, Z.; Salavati, M.; Buggiotti, L.; Takeda, H.; Tang, L.; Becker, F.; Ingvartsen, K.I.; Ferris, C.; Hostens, M.; et al. Relationships between Metabolic Profiles and Gene Expression in Liver and Leukocytes of Dairy Cows in Early Lactation. J. Dairy Sci. 2021, 104, 3596–3616. [Google Scholar] [CrossRef] [PubMed]
  71. Linke, V.; Overmyer, K.A.; Miller, I.J.; Brademan, D.R.; Hutchins, P.D.; Trujillo, E.A.; Reddy, T.R.; Russell, J.D.; Cushing, E.M.; Schueler, K.L.; et al. A Large-Scale Genome–Lipid Association Map Guides Lipid Identification. Nat. Metab. 2020, 2, 1149–1162. [Google Scholar] [CrossRef] [PubMed]
  72. Ghoreishifar, S.M.; Eriksson, S.; Johansson, A.M.; Khansefid, M.; Moghaddaszadeh-Ahrabi, S.; Parna, N.; Davoudi, P.; Javanmard, A. Signatures of Selection Reveal Candidate Genes Involved in Economic Traits and Cold Acclimation in Five Swedish Cattle Breeds. Genet. Sel. Evol. 2020, 52, 52. [Google Scholar] [CrossRef]
  73. Igoshin, A.V.; Yudin, N.S.; Belonogova, N.M.; Larkin, D.M. Genome-Wide Association Study for Body Weight in Cattle Populations from Siberia. Anim. Genet. 2019, 50, 250–253. [Google Scholar] [CrossRef] [PubMed]
  74. Hatzirodos, N.; Hummitzsch, K.; Irving-Rodgers, H.F.; Breen, J.; Perry, V.E.A.; Anderson, R.A.; Rodgers, R.J. Transcript Abundance of Stromal and Thecal Cell Related Genes during Bovine Ovarian Development. PLoS ONE 2019, 14, e0213575. [Google Scholar] [CrossRef]
  75. Kong, D.; He, M.; Yang, L.; Zhou, R.; Yan, Y.Q.; Liang, Y.; Teng, C.B. MiR-17 and MiR-19 Cooperatively Promote Skeletal Muscle Cell Differentiation. Cell. Mol. Life Sci. 2019, 76, 5041–5054. [Google Scholar] [CrossRef] [PubMed]
  76. Yoshioka, S.; Abe, H.; Sakumoto, R.; Okuda, K. Proliferation of Luteal Steroidogenic Cells in Cattle. PLoS ONE 2013, 8, e84186. [Google Scholar] [CrossRef]
  77. Aglan, H.S.; Gebremedhn, S.; Salilew-Wondim, D.; Neuhof, C.; Tholen, E.; Holker, M.; Schellander, K.; Tesfaye, D. Regulation of Nrf2 and NF-ΚB during Lead Toxicity in Bovine Granulosa Cells. Cell Tissue Res. 2020, 380, 643–655. [Google Scholar] [CrossRef] [PubMed]
  78. Baddela, V.S.; Sharma, A.; Michaelis, M.; Vanselow, J. HIF1 Driven Transcriptional Activity Regulates Steroidogenesis and Proliferation of Bovine Granulosa Cells. Sci. Rep. 2020, 10, 3906. [Google Scholar] [CrossRef]
  79. Dias, F.C.F.; Khan, M.I.R.; Sirard, M.A.; Adams, G.P.; Singh, J. Transcriptome Analysis of Granulosa Cells after Conventional vs Long FSH-Induced Superstimulation in Cattle. BMC Genom. 2018, 19, 258. [Google Scholar] [CrossRef]
  80. Gupta, P.S.P.; Folger, J.K.; Rajput, S.K.; Lv, L.; Yao, J.; Ireland, J.J.; Smith, G.W. Regulation and Regulatory Role of WNT Signaling in Potentiating FSH Action during Bovine Dominant Follicle Selection. PLoS ONE 2014, 9, e100201. [Google Scholar] [CrossRef] [PubMed]
  81. Sharma, A.; Baddela, V.S.; Roettgen, V.; Vernunft, A.; Viergutz, T.; Dannenberger, D.; Hammon, H.M.; Schoen, J.; Vanselow, J. Effects of Dietary Fatty Acids on Bovine Oocyte Competence and Granulosa Cells. Front. Endocrinol. 2020, 11, 87. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, W.; Zou, H.; Chen, N.; Tian, Y.; Hao, H.; Pang, Y.; Zhao, X.; Zhu, H.; Yu, D.; Du, W. Knockdown of CLAUDIN-6 Inhibited Apoptosis and Induced Proliferation of Bovine Cumulus Cells. Int. J. Mol. Sci. 2022, 23, 3222. [Google Scholar] [CrossRef] [PubMed]
  83. Xie, J.; Kalwar, Q.; Yan, P.; Guo, X. Effect of Concentrate Supplementation on the Expression Profile of Mirna in the Ovaries of Yak during Non-Breeding Season. Animals 2020, 10, 1640. [Google Scholar] [CrossRef] [PubMed]
  84. Yenuganti, V.R.; Viergutz, T.; Vanselow, J. Oleic Acid Induces Specific Alterations in the Morphology, Gene Expression and Steroid Hormone Production of Cultured Bovine Granulosa Cells. Gen. Comp. Endocrinol. 2016, 232, 134–144. [Google Scholar] [CrossRef]
  85. Wei, Y.; Cui, Y.F.; Tong, H.L.; Zhang, W.W.; Yan, Y.Q. MicroRNA-2400 Promotes Bovine Preadipocyte Proliferation. Biochem. Biophys. Res. Commun. 2016, 478, 1054–1059. [Google Scholar] [CrossRef]
  86. Du, J.; Zhao, H.; Song, G.; Pang, Y.; Jiang, L.; Zan, L.; Wang, H. Overexpression of Cholinergic Receptor Nicotinic Gamma Subunit Inhibits Proliferation and Differentiation of Bovine Preadipocytes. Anim. Biosci. 2023, 36, 200–208. [Google Scholar] [CrossRef]
  87. Zhou, D.; Wang, Y.; Yang, R.; Wang, F.; Zhao, Z.; Wang, X.; Xie, L.; Tian, X.; Wang, G.; Li, B.; et al. The MyoD1 Promoted Muscle Differentiation and Generation by Activating CCND2 in Guanling Cattle. Animals 2022, 12, 2571. [Google Scholar] [CrossRef]
  88. Jiang, J.; Cole, J.B.; Freebern, E.; Da, Y.; VanRaden, P.M.; Ma, L. Functional Annotation and Bayesian Fine-Mapping Reveals Candidate Genes for Important Agronomic Traits in Holstein Bulls. Commun. Biol. 2019, 2, 212. [Google Scholar] [CrossRef]
  89. An, B.; Xia, J.; Chang, T.; Wang, X.; Xu, L.; Zhang, L.; Gao, X.; Chen, Y.; Li, J.; Gao, H. Genome-Wide Association Study Reveals Candidate Genes Associated with Body Measurement Traits in Chinese Wagyu Beef Cattle. Anim. Genet. 2019, 50, 386–390. [Google Scholar] [CrossRef]
  90. Fang, L.; Cai, W.; Liu, S.; Canela-Xandri, O.; Gao, Y.; Jiang, J.; Rawlik, K.; Li, B.; Schroeder, S.G.; Rosen, B.D.; et al. Comprehensive Analyses of 723 Transcriptomes Enhance Genetic and Biological Interpretations for Complex Traits in Cattle. Genome Res. 2020, 30, 790–801. [Google Scholar] [CrossRef] [PubMed]
  91. Randell, A.; Daneshtalab, N. Elastin Microfibril Interface–Located Protein 1, Transforming Growth Factor Beta, and Implications on Cardiovascular Complications. J. Am. Soc. Hypertens. 2017, 11, 437–448. [Google Scholar] [CrossRef] [PubMed]
  92. Bot, S.; Andreuzzi, E.; Capuano, A.; Schiavinato, A.; Colombatti, A.; Doliana, R. Multiple-Interactions among EMILIN1 and EMILIN2 N- and C-Terminal Domains. Matrix Biol. 2015, 41, 44–55. [Google Scholar] [CrossRef]
  93. Capuano, A.; Bucciotti, F.; Farwell, K.D.; Tippin Davis, B.; Mroske, C.; Hulick, P.J.; Weissman, S.M.; Gao, Q.; Spessotto, P.; Colombatti, A.; et al. Diagnostic Exome Sequencing Identifies a Novel Gene, EMILIN1, Associated with Autosomal-Dominant Hereditary Connective Tissue Disease. Hum. Mutat. 2016, 37, 84–97. [Google Scholar] [CrossRef]
  94. Minuti, A.; Jahan, N.; Lopreiato, V.; Piccioli-Cappelli, F.; Bomba, L.; Capomaccio, S.; Loor, J.J.; Ajmone-Marsan, P.; Trevisi, E. Evaluation of Circulating Leukocyte Transcriptome and Its Relationship with Immune Function and Blood Markers in Dairy Cows during the Transition Period. Funct. Integr. Genom. 2020, 20, 293–305. [Google Scholar] [CrossRef]
  95. Pinto De Melo, T.; De Camargo, G.M.F.; De Albuquerque, L.G.; Carvalheiro, R. Genome-Wide Association Study Provides Strong Evidence of Genes Affecting the Reproductive Performance of Nellore Beef Cows. PLoS ONE 2017, 12, e0178551. [Google Scholar] [CrossRef]
  96. Yin, T.; König, S. Genome-Wide Associations and Detection of Potential Candidate Genes for Direct Genetic and Maternal Genetic Effects Influencing Dairy Cattle Body Weight at Different Ages. Genet. Sel. Evol. 2019, 51, 4. [Google Scholar] [CrossRef]
  97. Lindberg, K.; Olauson, H.; Amin, R.; Ponnusamy, A.; Goetz, R.; Taylor, R.F.; Mohammadi, M.; Canfield, A.; Kublickiene, K.; Larsson, T.E. Arterial Klotho Expression and FGF23 Effects on Vascular Calcification and Function. PLoS ONE 2013, 8, e60658. [Google Scholar] [CrossRef]
  98. Mattinzoli, D.; Rastaldi, M.P.; Ikehata, M.; Armelloni, S.; Pignatari, C.; Giardino, L.A.; Li, M.; Alfieri, C.M.; Regalia, A.; Riccardi, D.; et al. FGF23-Regulated Production of Fetuin-A (AHSG) in Osteocytes. Bone 2016, 83, 35–47. [Google Scholar] [CrossRef]
  99. Blau, J.E.; Collins, M.T. The PTH-Vitamin D-FGF23 Axis. Rev. Endocr. Metab. Disord. 2015, 16, 165–174. [Google Scholar] [CrossRef]
  100. Imel, E.A.; Biggin, A.; Schindeler, A.; Munns, C.F. FGF23, Hypophosphatemia, and Emerging Treatments. JBMR Plus 2019, 3, e10190. [Google Scholar] [CrossRef] [PubMed]
  101. Kinoshita, Y.; Fukumoto, S. X-Linked Hypophosphatemia and FGF23-Related Hypophosphatemic Diseases: Prospect for New Treatment. Endocr. Rev. 2018, 39, 274–291. [Google Scholar] [CrossRef] [PubMed]
  102. Onal, M.; Carlson, A.H.; Thostenson, J.D.; Benkusky, N.A.; Meyer, M.B.; Lee, S.M.; Pike, J.W. A Novel Distal Enhancer Mediates Inflammation-, PTH-, and Early Onset Murine Kidney Disease-Induced Expression of the Mouse Fgf23 Gene. JBMR Plus 2018, 2, 31–46. [Google Scholar] [CrossRef] [PubMed]
  103. Bernard, C.; Cassar-Malek, I.; Renand, G.; Hocquette, J.F. Changes in Muscle Gene Expression Related to Metabolism According to Growth Potential in Young Bulls. Meat Sci. 2009, 82, 205–212. [Google Scholar] [CrossRef]
  104. Shamsi, F.; Xue, R.; Huang, T.L.; Lundh, M.; Liu, Y.; Leiria, L.O.; Lynes, M.D.; Kempf, E.; Wang, C.H.; Sugimoto, S.; et al. FGF6 and FGF9 Regulate UCP1 Expression Independent of Brown Adipogenesis. Nat. Commun. 2020, 11, 1421. [Google Scholar] [CrossRef] [PubMed]
  105. Cooke, F.N.T.; Pennington, K.A.; Yang, Q.; Ealy, A.D. Several Fibroblast Growth Factors Are Expressed during Pre-Attachment Bovine Conceptus Development and Regulate Interferon-Tau Expression from Trophectoderm. Reproduction 2009, 137, 259–269. [Google Scholar] [CrossRef] [PubMed]
  106. Campos, C.; Valente, L.M.P.; Borges, P.; Bizuayehu, T.; Fernandes, J.M.O. Dietary Lipid Levels Have a Remarkable Impact on the Expression of Growth-Related Genes in Senegalese Sole (Solea senegalensis Kaup). J. Exp. Biol. 2010, 213, 200–209. [Google Scholar] [CrossRef]
  107. Xu, Y.; Tan, Q.; Hu, P.; Yao, J. Characterization and Expression Analysis of FGF6 (Fibroblast Growth Factor 6) Genes of Grass Carp (Ctenopharyngodon idellus) Reveal Their Regulation on Muscle Growth. Fish Physiol. Biochem. 2019, 45, 1649–1662. [Google Scholar] [CrossRef]
  108. Armand, A.S.; Laziz, I.; Chanoine, C. FGF6 in Myogenesis. Biochim. Biophys. Acta Mol. Cell Res. 2006, 1763, 773–778. [Google Scholar] [CrossRef]
  109. Laziz, I.; Armand, A.S.; Pariset, C.; Lecolle, S.; Della Gaspera, B.; Charbonnier, F.; Chanoine, C. Sprouty Gene Expression Is Regulated by Nerve and FGF6 during Regeneration of Mouse Muscles. Growth Factors 2007, 25, 151–159. [Google Scholar] [CrossRef]
  110. Fortes, M.R.S.; Reverter, A.; Kelly, M.; Mcculloch, R.; Lehnert, S.A. Genome-Wide Association Study for Inhibin, Luteinizing Hormone, Insulin-like Growth Factor 1, Testicular Size and Semen Traits in Bovine Species. Andrology 2013, 1, 644–650. [Google Scholar] [CrossRef] [PubMed]
  111. Hazeslip, L.; Zafar, M.K.; Chauhan, M.Z.; Byrd, A.K. Genome Maintenance by DNA Helicase B. Genes 2020, 11, 578. [Google Scholar] [CrossRef] [PubMed]
  112. Sun, L.; Qu, K.; Ma, X.; Hanif, Q.; Zhang, J.; Liu, J.; Chen, N.; Suolang, Q.; Lei, C.; Huang, B. Whole-Genome Analyses Reveal Genomic Characteristics and Selection Signatures of Lincang Humped Cattle at the China–Myanmar Border. Front. Genet. 2022, 13, 833503. [Google Scholar] [CrossRef] [PubMed]
  113. Maiorano, A.M.; Cardoso, D.F.; Carvalheiro, R.; Júnior, G.A.F.; de Albuquerque, L.G.; de Oliveira, H.N. Signatures of Selection in Nelore Cattle Revealed by Whole-Genome Sequencing Data. Genomics 2022, 114, 110304. [Google Scholar] [CrossRef] [PubMed]
  114. Melnik, B.C.; Schmitz, G. Milk Exosomal MicroRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int. J. Mol. Sci. 2022, 23, 11503. [Google Scholar] [CrossRef]
  115. Nair, R.; Gupta, P.; Shanmugam, M. Mitochondrial Metabolic Determinants of Multiple Myeloma Growth, Survival, and Therapy Efficacy. Front. Oncol. 2022, 12, 1000106. [Google Scholar] [CrossRef]
  116. Saatchi, M.; Schnabel, R.D.; Taylor, J.F.; Garrick, D.J. Large-Effect Pleiotropic or Closely Linked QTL Segregate within and across Ten US Cattle Breeds. BMC Genom. 2014, 15, 442. [Google Scholar] [CrossRef]
  117. Kim, S.J.; Ka, S.; Ha, J.W.; Kim, J.; Yoo, D.A.; Kim, K.; Lee, H.K.; Lim, D.; Cho, S.; Hanotte, O.; et al. Cattle Genome-Wide Analysis Reveals Genetic Signatures in Trypanotolerant N’Dama. BMC Genom. 2017, 18, 371. [Google Scholar] [CrossRef]
  118. Bensaad, K.; Cheung, E.C.; Vousden, K.H. Modulation of Intracellular ROS Levels by TIGAR Controls Autophagy. EMBO J. 2009, 28, 3015–3026. [Google Scholar] [CrossRef]
  119. Tang, J.; Chen, L.; Qin, Z.-H.; Sheng, R. Structure, Regulation, and Biological Functions of TIGAR and Its Role in Diseases. Acta Pharmacol. Sin. 2021, 42, 1547–1555. [Google Scholar] [CrossRef]
  120. Marete, A.; Sahana, G.; Fritz, S.; Lefebvre, R.; Barbat, A.; Lund, M.S.; Guldbrandtsen, B.; Boichard, D. Genome-Wide Association Study for Milking Speed in French Holstein Cows. J. Dairy Sci. 2018, 101, 6205–6219. [Google Scholar] [CrossRef] [PubMed]
  121. Simon-Molas, H.; Sánchez-De-Diego, C.; Navarro-Sabaté, À.; Castaño, E.; Ventura, F.; Bartrons, R.; Manzano, A. The Expression of TP53-Induced Glycolysis and Apoptosis Regulator (TIGAR) Can Be Controlled by the Antioxidant Orchestrator NRF2 in Human Carcinoma Cells. Int. J. Mol. Sci. 2022, 23, 1905. [Google Scholar] [CrossRef] [PubMed]
  122. Chebii, V.J.; Mpolya, E.A.; Muchadeyi, F.C.; Entfellner, J.B.D. Genomics of Adaptations in Ungulates. Animals 2021, 11, 1617. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, Y.; Ji, N.; Wang, J.; Cao, J.; Li, D.; Zhang, Y.; Zhang, L. SCG3 Protein Expression in Glioma Associates with Less Malignancy and Favorable Clinical Outcomes. Pathol. Oncol. Res. 2021, 27, 594931. [Google Scholar] [CrossRef] [PubMed]
  124. Dai, C.; Tian, H.; Bhatt, A.; Su, G.; Webster, K.A.; Li, W. Safety and Efficacy of Systemic Anti-Scg3 Therapy to Treat Oxygen-Induced Retinopathy. Front. Biosci.-Landmark 2022, 27, 130. [Google Scholar] [CrossRef]
  125. Tanabe, A.; Yanagiya, T.; Iida, A.; Saito, S.; Sekine, A.; Takahashi, A.; Nakamura, T.; Tsunoda, T.; Kamohara, S.; Nakata, Y.; et al. Functional Single-Nucleotide Polymorphisms in the Secretogranin III (SCG3) Gene That Form Secretory Granules with Appetite-Related Neuropeptides Are Associated with Obesity. J. Clin. Endocrinol. Metab. 2007, 92, 1145–1154. [Google Scholar] [CrossRef]
  126. Dias, M.M.; Cánovas, A.; Mantilla-Rojas, C.; Riley, D.G.; Luna-Nevarez, P.; Coleman, S.J.; Speidel, S.E.; Enns, R.M.; Islas-Trejo, A.; Medrano, J.F.; et al. SNP Detection Using RNA-Sequences of Candidate Genes Associated with Puberty in Cattle. Genet. Mol. Res. 2017, 16, gmr16019522. [Google Scholar] [CrossRef]
  127. Keogh, K.; Kenny, D.A. Gene Co-Expression Networks Contributing to Reproductive Development in Holstein-Friesian Bull Calves. Animal 2022, 16, 100527. [Google Scholar] [CrossRef]
  128. Sánchez, J.M.; Keogh, K.; Kelly, A.K.; Byrne, C.J.; Lonergan, P.; Kenny, D.A. A High Plane of Nutrition during Early Life Alters the Hypothalamic Transcriptome of Heifer Calves. Sci. Rep. 2021, 11, 13978. [Google Scholar] [CrossRef]
  129. Lin, S.; Fang, L.; Kang, X.; Liu, S.; Liu, M.; Connor, E.E.; Baldwin, R.L.; Liu, G.; Li, C.J. Establishment and Transcriptomic Analyses of a Cattle Rumen Epithelial Primary Cells (REPC) Culture by Bulk and Single-Cell RNA Sequencing to Elucidate Interactions of Butyrate and Rumen Development. Heliyon 2020, 6, e04112. [Google Scholar] [CrossRef]
  130. Kano, C.; Hanaoka, F.; Wang, J.Y. Analysis of Mice Deficient in Both REV1 Catalytic Activity and POLH Reveals an Unexpected Role for POLH in the Generation of C to g and G to C Transversions during Ig Gene Hypermutation. Int. Immunol. 2012, 24, 169–174. [Google Scholar] [CrossRef] [PubMed]
  131. Masuda, K.; Ouchida, R.; Hikida, M.; Kurosaki, T.; Yokoi, M.; Masutani, C.; Seki, M.; Wood, R.D.; Hanaoka, F.; O-Wang, J. DNA Polymerases η and θ Function in the Same Genetic Pathway to Generate Mutations at A/T during Somatic Hypermutation of Ig Genes. J. Biol. Chem. 2007, 282, 17387–17394. [Google Scholar] [CrossRef]
  132. Roerink, S.F.; Koole, W.; Stapel, L.C.; Romeijn, R.J.; Tijsterman, M. A Broad Requirement for TLS Polymerases η and κ, and Interacting Sumoylation and Nuclear Pore Proteins, in Lesion Bypass during C. Elegans Embryogenesis. PLoS Genet. 2012, 8, e1002800. [Google Scholar] [CrossRef] [PubMed]
  133. Giotti, B.; Chen, S.H.; Barnett, M.W.; Regan, T.; Ly, T.; Wiemann, S.; Hume, D.A.; Freeman, T.C. Assembly of a Parts List of the Human Mitotic Cell Cycle Machinery. J. Mol. Cell Biol. 2019, 11, 703–718. [Google Scholar] [CrossRef] [PubMed]
  134. Diepeveen, E.T.; Roth, O.; Salzburger, W. Immune-Related Functions of the Hivep Gene Family in East African Cichlid Fishes. G3 Genes. Genomes Genet. 2013, 3, 2205–2217. [Google Scholar] [CrossRef]
  135. Dürr, U.; Henningfeld, K.A.; Hollemann, T.; Knöchel, W.; Pieler, T. Isolation and Characterization of the Xenopus HIVEP Gene Family. Eur. J. Biochem. 2004, 271, 1135–1144. [Google Scholar] [CrossRef] [PubMed]
  136. Zare, Y.; Shook, G.E.; Collins, M.T.; Kirkpatrick, B.W. Genome-Wide Association Analysis and Genomic Prediction of Mycobacterium Avium Subspecies Paratuberculosis Infection in US Jersey Cattle. PLoS ONE 2014, 9, e88380. [Google Scholar] [CrossRef] [PubMed]
  137. Palombo, V.; D’Andrea, M.; Licastro, D.; Monego, S.D.; Sgorlon, S.; Sandri, M.; Stefanon, B. Single-Step Genome Wide Association Study Identifies Qtl Signals for Untrimmed and Trimmed Thigh Weight in Italian Crossbred Pigs for Dry-Cured Ham Production. Animals 2021, 11, 1612. [Google Scholar] [CrossRef]
  138. Kang, H.-C.; Kim, H.-S.; Lee, J.-B.; Yoo, C.-K.; Choi, T.-J.; Lim, H.-T. Analysis of the Relationship between the End Weight Trait and the Gene ADGRL2 in Purebred Landrace Pigs Using a Genome-Wide Association Study. Korean J. Agric. Sci. 2018, 45, 238–247. [Google Scholar] [CrossRef]
  139. Lagou, V.; Garcia-Perez, J.E.; Smets, I.; Van Horebeek, L.; Vandebergh, M.; Chen, L.; Mallants, K.; Prezzemolo, T.; Hilven, K.; Humblet-Baron, S.; et al. Genetic Architecture of Adaptive Immune System Identifies Key Immune Regulators. Cell Rep. 2018, 25, 798–810.e6. [Google Scholar] [CrossRef]
  140. Ueda, S.; Hosoda, M.; Yoshino, K.-I.; Yamanoue, M.; Shirai, Y. Gene Expression Analysis Provides New Insights into the Mechanism of Intramuscular Fat Formation in Japanese Black Cattle. Genes 2021, 12, 1107. [Google Scholar] [CrossRef] [PubMed]
  141. Meuwissen, M.E.C.; Halley, D.J.J.; Smit, L.S.; Lequin, M.H.; Cobben, J.M.; De Coo, R.; Van Harssel, J.; Sallevelt, S.; Woldringh, G.; Van Der Knaap, M.S.; et al. The Expanding Phenotype of COL4A1 and COL4A2 Mutations: Clinical Data on 13 Newly Identified Families and a Review of the Literature. Genet. Med. 2015, 17, 843–853. [Google Scholar] [CrossRef]
  142. Pollner, R.; Schmidt, C.; Fischer, G.; Kühn, K.; Pöschl, E. Cooperative and Competitive Interactions of Regulatory Elements Are Involved in the Control of Divergent Transcription of Human Col4Al and Col4A2 Genes. FEBS Lett. 1997, 405, 31–36. [Google Scholar] [CrossRef] [PubMed]
  143. Kuo, D.S.; Labelle-Dumais, C.; Gould, D.B. Col4a1 and Col4a2 Mutations and Disease: Insights into Pathogenic Mechanisms and Potential Therapeutic Targets. Hum. Mol. Genet. 2012, 21, R97–R110. [Google Scholar] [CrossRef]
  144. Suárez-Vega, A.; Gutiérrez-Gil, B.; Klopp, C.; Robert-Granie, C.; Tosser-Klopp, G.; Arranz, J.J. Characterization and Comparative Analysis of the Milk Transcriptome in Two Dairy Sheep Breeds Using RNA Sequencing. Sci. Rep. 2015, 5, 18399. [Google Scholar] [CrossRef]
  145. Dai, W.; Chen, Q.; Wang, Q.; White, R.R.; Liu, J.; Liu, H. Complementary Transcriptomic and Proteomic Analyses Reveal Regulatory Mechanisms of Milk Protein Production in Dairy Cows Consuming Different Forages. Sci. Rep. 2017, 7, 44234. [Google Scholar] [CrossRef] [PubMed]
  146. Dai, W.; Wang, Q.; Zhao, F.; Liu, J.; Liu, H. Understanding the Regulatory Mechanisms of Milk Production Using Integrative Transcriptomic and Proteomic Analyses: Improving Inefficient Utilization of Crop by-Products as Forage in Dairy Industry. BMC Genom. 2018, 19, 403. [Google Scholar] [CrossRef]
  147. Bai, Y.; Li, X.; Chen, Z.; Li, J.; Tian, H.; Ma, Y.; Raza, S.H.A.; Shi, B.; Han, X.; Luo, Y.; et al. Interference with ACSL1 Gene in Bovine Adipocytes: Transcriptome Profiling of MRNA and LncRNA Related to Unsaturated Fatty Acid Synthesis. Front. Vet. Sci. 2021, 8, 788316. [Google Scholar] [CrossRef]
  148. Liu, R.; Liu, X.; Bai, X.; Xiao, C.; Dong, Y. Different Expression of Lipid Metabolism-Related Genes in Shandong Black Cattle and Luxi Cattle Based on Transcriptome Analysis. Sci. Rep. 2020, 10, 21915. [Google Scholar] [CrossRef]
  149. Bryckaert, M.; Guillonneau, X.; Hecquet, C.; Perani, P.; Courtois, Y.; Mascarelli, F. Regulation of Proliferation-Survival Decisions Is Controlled by FGF1 Secretion in Retinal Pigmented Epithelial Cells. Oncogene 2000, 19, 4917–4929. [Google Scholar] [CrossRef]
  150. Hsu, Y.C.; Kao, C.Y.; Chung, Y.F.; Chen, M.S.; Chiu, I.M. Ciliogenic RFX Transcription Factors Regulate Fgf1 Gene Promoter. J. Cell Biochem. 2012, 113, 2511–2522. [Google Scholar] [CrossRef]
  151. Uchida, S.; Teubner, B.J.W.; Hevi, C.; Hara, K.; Kobayashi, A.; Dave, R.M.; Shintaku, T.; Jaikhan, P.; Yamagata, H.; Suzuki, T.; et al. CRTC1 Nuclear Translocation Following Learning Modulates Memory Strength via Exchange of Chromatin Remodeling Complexes on the Fgf1 Gene. Cell Rep. 2017, 18, 352–366. [Google Scholar] [CrossRef]
  152. Butt, C.; Lim, S.; Greenwood, C.; Rahman, P. VEGF, FGF1, FGF2 and EGF Gene Polymorphisms and Psoriatic Arthritis. BMC Musculoskelet. Disord. 2007, 8, 1. [Google Scholar] [CrossRef] [PubMed]
  153. Einspanier, R.; Kettler, A.; Gabler, C.; Kloas, W.; Einspanier, A.; Schams, D. The Mammalian Oviduct: Aspects on Auto-and Paracrine Mechanisms. Reprod. Domest. Anim. 2000, 35, 125–128. [Google Scholar] [CrossRef]
  154. Han, P.; Guerrero-Netro, H.; Estienne, A.; Cao, B.; Price, C.A. Regulation and Action of Early Growth Response 1 in Bovine Granulosa Cells. Reproduction 2017, 154, 547–557. [Google Scholar] [CrossRef] [PubMed]
  155. Herzog, K.; Debertolis, L.; Kastelic, J.P.; Schmicke, M.; Ulbrich, S.E.; Bollwein, H. Effects of Intravenous Infusion of E. Coli Lipopolysaccharide in Early Pregnant Cows. Reproduction 2019, 157, 65–76. [Google Scholar] [CrossRef] [PubMed]
  156. Jiang, Z.; Price, C.A. Differential Actions of Fibroblast Growth Factors on Intracellular Pathways and Target Gene Expression in Bovine Ovarian Granulose Cells. Reproduction 2012, 144, 625–632. [Google Scholar] [CrossRef]
  157. Pfarrer, C.; Weise, S.; Berisha, B.; Schams, D.; Leiser, R.; Hoffmann, B.; Schuler, G. Fibroblast Growth Factor (FGF)-1, FGF2, FGF7 and FGF Receptors Are Uniformly Expressed in Trophoblast Giant Cells during Restricted Trophoblast Invasion in Cows. Placenta 2006, 27, 758–770. [Google Scholar] [CrossRef]
  158. Zhou, Y.; Ren, L.; Zhu, J.; Yan, S.; Wang, H.; Song, N.; Li, L.; Ouyang, H.; Pang, D. Construction of a Recombinant Human FGF1 Expression Vector for Mammary Gland-Specific Expression in Human Breast Cancer Cells. Mol. Cell Biochem. 2011, 354, 39–46. [Google Scholar] [CrossRef]
  159. Bergfelder-Drüing, S.; Grosse-Brinkhaus, C.; Lind, B.; Erbe, M.; Schellander, K.; Simianer, H.; Tholen, E. A Genome-Wide Association Study in Large White and Landrace Pig Populations for Number Piglets Born Alive. PLoS ONE 2015, 10, e0117468. [Google Scholar] [CrossRef]
  160. Picking, E.M.; Trubenbach, L.A.B.F.W.; Sawyer, J.E.; Wickersham, T.A.; Satterfield, M. Carey Technical Note: Relationship between Placentome Location and Gene Expression in Bovine Pregnancy. J. Anim. Sci. 2020, 98, skaa176. [Google Scholar] [CrossRef] [PubMed]
  161. Rotta, P.P.; Valadares Filho, S.C.; Gionbelli, T.R.S.; Costa e Silva, L.F.; Engle, T.E.; Marcondes, M.I.; Guimarães, S.E.F.; Nascimento, C.S.; Carvalho, B.C.; Silva, F.A.S.; et al. Effects of Day of Gestation and Feeding Regimen in Holstein × Gyr Cows: III. Placental Adaptations and Placentome Gene Expression. J. Dairy Sci. 2015, 98, 3224–3235. [Google Scholar] [CrossRef]
  162. Saǧsöz, H.; Saruhan, B.G. The Expression of Vascular Endothelial Growth Factor and Its Receptors (Flt1/Fms, Flk1/KDR, Flt4) and Vascular Endothelial Growth Inhibitor in the Bovine Uterus during the Sexual Cycle and Their Correlation with Serum Sex Steroids. Theriogenology 2011, 75, 1720–1734. [Google Scholar] [CrossRef] [PubMed]
  163. Liu, D.; Chen, Z.; Zhao, W.; Guo, L.; Sun, H.; Zhu, K.; Liu, G.; Shen, X.; Zhao, X.; Wang, Q.; et al. Genome-Wide Selection Signatures Detection in Shanghai Holstein Cattle Population Identified Genes Related to Adaption, Health and Reproduction Traits. BMC Genom. 2021, 22, 747. [Google Scholar] [CrossRef] [PubMed]
  164. Yin, T.; Halli, K.; König, S. Direct Genetic Effects, Maternal Genetic Effects, and Maternal Genetic Sensitivity on Prenatal Heat Stress for Calf Diseases and Corresponding Genomic Loci in German Holsteins. J. Dairy Sci. 2022, 105, 6795–6808. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Manhattan plots displaying the association of genomic markers with Final Classification (FC), Dairy Strength (DS), Rump, Feet and Legs (FL), and Mammary System (MS) traits in Brazilian Holstein cattle. The red line indicates the significant threshold for the association with the trait.
Figure 1. Manhattan plots displaying the association of genomic markers with Final Classification (FC), Dairy Strength (DS), Rump, Feet and Legs (FL), and Mammary System (MS) traits in Brazilian Holstein cattle. The red line indicates the significant threshold for the association with the trait.
Animals 14 02472 g001aAnimals 14 02472 g001bAnimals 14 02472 g001c
Figure 2. Gene interaction network for the genes associated with conformation traits in Brazilian Holstein cattle.
Figure 2. Gene interaction network for the genes associated with conformation traits in Brazilian Holstein cattle.
Animals 14 02472 g002
Table 1. Descriptive statistics for Final Classification (FC), Dairy Strength (DS), Rump, Feet and Legs (FL), and Mammary System (MS) in Brazilian Holstein cows.
Table 1. Descriptive statistics for Final Classification (FC), Dairy Strength (DS), Rump, Feet and Legs (FL), and Mammary System (MS) in Brazilian Holstein cows.
VariableNMean±SDMinimumMaximum
FC2339101.353.2189111
DS2339100.932.5988108
Rump233999.053.5782109
FL233998.933.1188110
MS2339103.874.3688119
N—number of animals in the analyses; SD—standard deviation.
Table 2. Description of the significant genomic markers, candidate genes, and number of QTLs significantly associated with Final Classification (FC), Dairy Strength (DS), Rump, Feet and Legs (FL), and Mammary System (MS) traits in Brazilian Holstein cattle.
Table 2. Description of the significant genomic markers, candidate genes, and number of QTLs significantly associated with Final Classification (FC), Dairy Strength (DS), Rump, Feet and Legs (FL), and Mammary System (MS) traits in Brazilian Holstein cattle.
TraitCHRPosition (bp)−log10
(p-Value)
Genes (±100 kb)QTLs
FC1058,643,6038.02TMOD2, TMOD3, LYSMD2, SCG3, DMXL2, ENSBTAG00000050593-
1058,635,4627.38TMOD2, TMOD3, LYMSD2, SCG3, ENSBTAG00000050593-
2644,637,0348.22ZRANB1, CTBP27
DS363,243,3347.55ADGRL2-
5112,981,4577.75SREBF2, bta-mir-33a, SHISA8, TNFRSF13C, CENPM, ENSBTAG00000050336, SEPTIN3, WBP2NL, NAGA, PHETA2, SMDT1, NDUFA6, MGC12705547
1163,571,1388.61RAB1A, ACTR215
1231,640,0208.54FLT1-
195,726,3867.54MMD1
222,988,2847.73ZCWPW22
2535,085,0417.46CUX1, MYL107
Rump547,431,1588.62GRIP1, U1, HELB, ENSBTAG0000005341923
5105,666,1597.72RAD51AP1, C5H12orf4, FGF6, FGF23, TIGAR, CCND227
1093,552,2727.60ENSBTAG00000050021-
1172,567,4627.82TCF23, PRR30, ABHD1, PREB, CGREF1, KHK, EMILIN1, AGBL5, TMEM214, MAPRE3, SNORA6215
1163,571,1387.64RAB1A, ACTR2
195,726,3867.77MMD1
2232,355,1697.91FRMD4B, LMOD3, ARL6IP51
FL363,219,3957.61ADGRL2-
97,763,0438.07ADGRB33
1461,736,1129.26ENSBTAG00000052148, KLF1027
226,375,5078.14OSBPL1015
2317,018,4647.91ZNF318, ABCC10, DLK2, TJAP1, LRRC73, YIPF3, POLR1C, XPO5, POLH16
2344,473,9427.41HIVEP1
MS753,622,2197.91FGF1, bta-mir-2460, ENSBTAG000000426245
1178,083,4137.60LDAH20
1285,218,2627.54COL4A2, RAB20, NAXD, CARS2, ING11
2814,058,0169.36BICC1-
Table 3. Gene ontology terms related to the genes associated with Final Classification (FC), Rump, Feet and Legs (FL), and Mammary System (MS) in Brazilian Holstein cattle.
Table 3. Gene ontology terms related to the genes associated with Final Classification (FC), Rump, Feet and Legs (FL), and Mammary System (MS) in Brazilian Holstein cattle.
TraitGOTermp-ValueGenes
FCGO:0051694Pointed-End Actin Filament Capping0.002TMOD2, TMOD3
GO:0030239Myofibril Assembly0.003TMOD2, TMOD3
GO:0006936Muscle Contraction0.013TMOD2, TMOD3
GO:0007015Actin Filament Organization0.038TMOD2, TMOD3
GO:0005865Striated Muscle Thin Filament0.002TMOD2, TMOD3
GO:0030016Myofibril0.005TMOD2, TMOD3
GO:0005856Cytoskeleton0.074TMOD2, TMOD3
GO:0005523Tropomyosin Binding0.002TMOD2, TMOD3
RumpGO:0010628Positive Regulation of Gene Expression0.006EMILIN1, FGF23, FGF6, TCF23
GO:1905168Positive Regulation of Double-Strand Break Repair via Homologous Recombination0.020RAD51AP1, ACTR2
GO:0045737Positive Regulation of Cyclin-Dependent Protein Serine/Threonine Kinase Activity0.024CCND2, MAPRE3
GO:0008543Fibroblast Growth Factor Receptor Signaling Pathway0.050FGF23, FGF6
GO:0008284Positive Regulation of Cell Proliferation0.059CCND2, FGF23, FGF6
GO:0030154Cell Differentiation0.083FGF23, FGF6, TCF23
GO:0009887Animal Organ Morphogenesis0.092FGF23, FGF6
GO:0005737Cytoplasm0.027HELB, FRMD4B, TIGAR, ACTR2, C5H12orf4, CCND2, FGF23, FGF6, KHK
GO:0005856Cytoskeleton0.040ARL6IP5, FRMD4B, LMOD3
GO:0005881Cytoplasmic Microtubule0.049MAPRE3, TMEM214
GO:0035861Site of Double-Strand Break0.067HELB, ACTR2
GO:0005104Fibroblast Growth Factor Receptor Binding0.022FGF23, FGF6
FLGO:0016020Membrane0.021ABCC10, ADGRL2, OSBPL10, TJAP1
GO:0005829Cytosol0.076POLH, HIVEP1, XPO5, OSBPL10, ZNF318
MSGO:0031012Extracellular Matrix0.066COL4A2, FGF1
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

Silva, E.F.P.; Gaia, R.C.; Mulim, H.A.; Pinto, L.F.B.; Iung, L.H.S.; Brito, L.F.; Pedrosa, V.B. Genome-Wide Association Study of Conformation Traits in Brazilian Holstein Cattle. Animals 2024, 14, 2472. https://doi.org/10.3390/ani14172472

AMA Style

Silva EFP, Gaia RC, Mulim HA, Pinto LFB, Iung LHS, Brito LF, Pedrosa VB. Genome-Wide Association Study of Conformation Traits in Brazilian Holstein Cattle. Animals. 2024; 14(17):2472. https://doi.org/10.3390/ani14172472

Chicago/Turabian Style

Silva, Emanueli F. P., Rita C. Gaia, Henrique A. Mulim, Luís Fernando Batista Pinto, Laiza H. S. Iung, Luiz F. Brito, and Victor B. Pedrosa. 2024. "Genome-Wide Association Study of Conformation Traits in Brazilian Holstein Cattle" Animals 14, no. 17: 2472. https://doi.org/10.3390/ani14172472

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop