Next Article in Journal
Analysis of the Microbial Community Structure of Ixodes persulcatus at Each Developmental Stage
Previous Article in Journal
Effects of Low-Protein Diet Without Soybean Meal on Growth Performance, Nutrient Digestibility, Plasma Free Amino Acids, and Meat Quality of Finishing Pigs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Leveraging Whole-Genome Resequencing to Uncover Genetic Diversity and Promote Conservation Strategies for Ruminants in Asia

Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
*
Authors to whom correspondence should be addressed.
Animals 2025, 15(6), 831; https://doi.org/10.3390/ani15060831
Submission received: 24 January 2025 / Revised: 28 February 2025 / Accepted: 12 March 2025 / Published: 13 March 2025
(This article belongs to the Section Animal Genetics and Genomics)

Simple Summary

Whole-genome resequencing is a powerful genomic technology for high-resolution identification of genetic variations. In ruminants, it has opened new research frontiers. It is widely used to study genetic diversity, helping understand evolutionary history, population structure, and relationships, which is crucial for conserving endangered breeds and genetic resources. For production performance, it has facilitated the discovery of genes related to milk yield, meat quality, and feed efficiency. Breeders can use this info to select superior animals for more efficient and sustainable farming. The progress in this technology holds great promise for advancing understanding of ruminant biology and improving farming productivity and sustainability.

Abstract

Whole-genome resequencing (WGRS) is a critical branch of whole-genome sequencing (WGS), primarily targeting species with existing reference genomes. By aligning sequencing data to the reference genome, WGRS enables precise detection of genetic variations in individuals or populations. As a core technology in genomic research, WGS holds profound significance in ruminant studies. It not only reveals the intricate structure of ruminant genomes but also provides essential data for deciphering gene function, variation patterns, and evolutionary processes, thereby advancing the exploration of ruminant genetic mechanisms. However, WGS still faces several challenges, such as incomplete and inaccurate genome assembly, as well as the incomplete annotation of numerous unknown genes or gene functions. Although WGS can identify a vast number of genomic variations, the specific relationships between these variations and phenotypes often remain unclear, which limits its potential in functional studies and breeding applications. By performing WGRS on multiple samples, these assembly challenges can be effectively addressed, particularly in regions with high repeat content or complex structural variations. WGRS can accurately identify subtle variations among different individuals or populations and further elucidate their associations with specific traits, thereby overcoming the limitations of WGS and providing more precise genetic information for functional research and breeding applications. This review systematically summarizes the latest applications of WGRS in the analysis of ruminant genetic structures, genetic diversity, economic traits, and adaptive traits, while also discussing the challenges faced by this technology. It aims to provide a scientific foundation for the improvement and conservation of ruminant genetic resources.

1. Introduction

Ruminants are closely intertwined with human daily life. According to a report by the Food and Agriculture Organization of the United Nations (FAO, https://www.fao.org/faostat/en/#data/FBS (accessed on 24 February 2025), China’s beef production reached 6.78885 million tons in 2023, while mutton production (including goat and sheep meat) totaled 5.31402 million tons. The gross production value of raw milk (from cattle, buffalo, goats, and sheep) amounted to 20,313,291 thousand international dollars (I$), and wool production reached 1,415,246 thousand I$. These figures underscore the pivotal role of ruminants in the global food and related industries. With advancing technology, significant breakthroughs have been made in ruminant genome research.
On 23 April 2009, the USDA Agricultural Research Service and Baylor College of Medicine, leading a consortium of over 300 scientists from 25 countries, announced the completion of cattle genome sequencing using a Hereford cow as the reference sample, employing BAC clone sequencing and whole-genome shotgun sequencing strategies [1]. In 2012, a research team led by Professor Jianquan Liu sequenced the genome of a female yak using whole-genome shotgun sequencing and the Illumina HiSeq 2000 platform, producing a 2657 Mb draft genome [2]. In 2017, a team from Northwest A&F University sequenced the genome of a Marco Polo sheep using whole-genome shotgun sequencing and the Illumina HiSeq 2000 platform, generating 1022.43 Gb of raw reads [3]. In 2019, researchers from the University of Adelaide constructed a high-resolution genome map of a female Mediterranean buffalo using single-molecule sequencing and chromatin conformation capture [4]. In 2024, Professor Menghua Li’s team at China Agricultural University assembled the first complete goat genome (T2T-goat1.0), spanning 2.86 Gb and including 20.96 Mb of the Y chromosome, using PacBio HiFi, Ultra-long ONT, Bionano, and Hi-C technologies [5]. With advancements in sequencing technologies, reduced costs, and improved assembly methods, high-quality genome sequences of an increasing number of species have been published.
With the continuous advancement of whole-genome sequencing (WGS) technologies, researchers are now able to obtain high-quality and complete reference genomes, which has laid a solid foundation for whole-genome resequencing (WGRS) studies in ruminants. By aligning WGRS data to reference genome sequences, this technology enables the detection of various genetic variants [6], identification of candidate genes associated with phenotypic traits [7], and analysis of population genetic diversity and structure [8,9]. Through resequencing analysis, researchers can deeply explore the genetic diversity of ruminants, precisely locate functional genes linked to desirable traits, and significantly accelerate the breeding process of new varieties. This approach plays a pivotal role in the development, utilization, and conservation of ruminant genetic resources.
This review summarizes recent advances in WGRS studies of ruminants and explores its applications in genetic resource conservation. The aim is to provide a scientific foundation for a deeper understanding of the genetic characteristics and diversity of ruminants, thereby promoting their conservation and sustainable development.

2. Overview of Whole-Genome Resequencing Technology

2.1. Definition

WGRS is a genomic sequencing technology designed for species with known genome sequences, enabling genomic analyses of individuals or populations to identify genetic differences. The core of this technology lies in the application of high-throughput sequencing to perform deep sequencing of genomes. Currently, the most widely used sequencing technologies are next-generation sequencing (NGS) and third-generation sequencing (TGS).
TGS is capable of generating long sequencing reads, typically based on single-molecule sequencing and sequencing in real-time principles, which directly read DNA or RNA molecules, thereby avoiding biases introduced by PCR amplification and improving detection accuracy [10]. This technology excels in identifying structural variations [11] and enables complete genome assembly [12]. In contrast, NGS is renowned for its high-throughput capabilities, featuring mature alignment and assembly algorithms, straightforward data quality control, and high cost-effectiveness [13]. In practical applications, TGS is more suitable for research objectives involving the resolution of complex genomic structures, investigation of gene integrity, detection of structural variations, or exploration of epigenetic modifications, leveraging its long-read advantages. On the other hand, NGS, with its short-read characteristics, is more advantageous for large-scale detection of single-nucleotide polymorphisms (SNPs) and insertions/deletions (InDels), screening of common genetic variants, and large-scale genomic sequencing projects where cost-effectiveness is a priority.
WGRS technology can detect a wide range of genetic variations, including SNPs, structural variations (SVs), copy number variations (CNVs), and InDels [14]. Depending on the research objective, WGRS can be categorized into individual-based and population-based approaches. Individual-based methods include high-coverage haplotype-unresolved whole-genome resequencing (huWGR) and high-coverage haplotype-resolved whole-genome resequencing (hrWGR). Population-based methods include pooled sample sequencing (Pool-seq) and low-coverage individual sequencing (lcWGR) [15].
WGRS has a wide range of applications, including the study of evolutionary processes [16,17], identification of functional genes, as well as its roles in animal and plant breeding [18,19], genetic map construction [20] and disease genomics research [21,22]. With the decreasing cost of sequencing and the development of bioinformatics tools, WGRS has become an efficient and cost-effective method for genomic analysis [23]. Its advantages lie in the comprehensive and precise acquisition of genomic information with high data output, enabling the discovery of numerous key genes associated with traits. As a result, WGRS has become an essential tool in modern biological research, driving advancements in multiple scientific fields.

2.2. Technical Advantages

WGRS, single-nucleotide polymorphism (SNP) arrays and WGS are pivotal technologies in genomics research, each offering distinct advantages and limitations in terms of resolution, cost-effectiveness, and applicability for studying genetic variations.
In recent years, WGRS has been widely applied in genomic research due to its distinct advantages, including high resolution, high coverage, cost-effectiveness, and low input requirements. The high resolution of WGRS stems from its ability to perform deep sequencing [24,25], advanced sequencing technologies [26], precise detection of genetic variations [27,28], and rigorous bioinformatics workflows [29]. These features make WGRS a powerful tool for studying genomic complexity and genetic diversity. Its high coverage capability, exemplified by single-molecule real-time sequencing (SMRT), significantly improves the assembly of high-GC-content genomes due to its unbiased sequencing and long-read lengths, enhancing coverage in GC-rich and repetitive regions [30]. Regarding cost-effectiveness, the continuous development of new sequencing platforms has led to a substantial reduction in sequencing costs while significantly increasing data output [31]. Furthermore, WGRS requires minimal input DNA, with starting amounts as low as picograms (pg) [32]. This low-input requirement is particularly advantageous for rare or clinical samples, allowing researchers to perform comprehensive genomic analyses with limited material.
SNP arrays are a crucial tool in genomics research, serving as a high-throughput genotyping technology designed to detect single-nucleotide polymorphisms (SNPs) at specific genomic loci [33]. By utilizing pre-designed probes, SNP arrays can simultaneously analyze thousands to millions of known SNP sites, enabling rapid and efficient genotyping of samples [34]. However, SNP arrays are limited to detecting only the pre-designed SNP loci on the chip, with a finite number of detectable sites [35]. They cannot identify SNPs outside the array or other types of genetic variations. In contrast, whole-genome resequencing (WGRS) sequences the entire genome, providing a comprehensive view of all genetic variations, including SNPs, InDels, CNVs, and SV. SNP arrays typically have a lower cost per sample, particularly when analyzing large sample sizes. They are a cost-effective choice when the research focus is on genotyping known SNPs and does not require whole-genome analysis [36]. On the other hand, WGRS involves extensive sequencing and data analysis, resulting in a higher cost per sample. However, with recent advancements in sequencing technologies, costs have significantly decreased. Moreover, for studies requiring the simultaneous analysis of multiple types of genetic variations, WGRS may prove more cost-effective in the long term compared to using multiple distinct methods to detect different types of variations separately [37].
Whole-genome sequencing (WGS) is a critical genomic tool that provides comprehensive genomic information. WGS involves the initial sequencing of the complete genome of a species or individual, aiming to construct a reference genome for the species [38]. Data interpretation relies on de novo assembly, which requires overcoming technical challenges such as repetitive sequences and complex genomic regions (e.g., high GC content or polyploid genomes) [39]. However, since WGS typically targets only a few individuals (or even a single individual), its limitation lies in its inability to directly reflect genetic diversity within populations. Although it provides essential foundational data for subsequent research, it struggles to deeply analyze genetic differences among individuals within a population or systematically compare genetic differentiation between populations. In contrast, WGRS effectively addresses these shortcomings. By aligning sequencing data to a reference genome, WGRS can accurately identify various types of genetic variations, including SNPs, InDels, and SVs [40]. This makes WGRS a powerful tool for studying population genetic structures, identifying functional genes, and conducting trait association analyses, thereby offering deeper insights into genetic differences and evolutionary mechanisms within and between populations.
In conclusion, WGRS, SNP arrays and WGS play indispensable roles in genomics research, each tailored to specific research needs and objectives. WGRS excels in providing comprehensive genomic insights, offering high resolution, extensive coverage, and the ability to detect a wide range of genetic variations, making it particularly valuable for studies requiring detailed genomic analysis and low-input samples.

3. Applications and Research Progress in Ruminants

3.1. Population Genetic Structure Analysis

Population genetic structure analysis in ruminants focuses on examining genetic differences among populations and understanding the historical dynamics of populations. By analyzing the genetic structure of these economically important animals, researchers can uncover their evolutionary history, population differentiation, and adaptation to diverse environmental conditions [41]. These insights are crucial for guiding population management and breeding programs, conserving genetic resources, and improving production performance and adaptability in ruminants.

3.1.1. Genetic Structure of Sheep and Goats

Recent studies on the genetic structure of sheep and goats have revealed genetic similarities, levels of differentiation, and population dynamics among various breeds.
Whole-genome sequencing of six Tibetan sheep breeds was performed using Illumina technology, and the obtained genomic data were aligned to the reference genome to identify two types of genetic variations, SNPs and InDels. Phylogenetic tree analysis based on the genetic variation data revealed genetic differentiation among these breeds, although the degree of divergence was relatively low, with extensive gene flow observed between populations. These sheep breeds were classified into three ecological types: plateau type, valley type, and Oula type [42]. Integrating genomic and transcriptomic analyses indicated that the Lanping black-boned sheep originated from the Lanping ordinary sheep and has evolved into a distinct breed [43]. A study that compared resequencing data from eight sheep breeds, involving 28 individuals, with the reference genome found that Tong sheep clustered with coarse-wool breeds such as Hu sheep, small-tailed Han sheep, Lop sheep, and Ujumqin sheep, while Hanzhong sheep showed greater genetic distance from Tong sheep. Principal component analysis (PCA) and ADMIXTURE results indicated that Tong sheep and other coarse-wool breeds belong to the Chinese Mongolian/Kazakh sheep lineage, whereas Hanzhong sheep maintained a relatively isolated breeding environment and genetic background due to its unique geographical location [44]. Further analysis of four sheep populations (European, Yunnan, Kazakh, and Mongolian) revealed that Tibetan sheep (GBF) clustered with Kazakh and Mongolian sheep in PCA but showed genetic contributions from Yunnan sheep in population structure analysis. Gene flow between Tibetan sheep and other populations was likely influenced by historical breeding practices and geographical proximity [45]. Studies have classified domestic sheep into African indigenous, Chinese indigenous, and improved breeds. Chinese indigenous breeds are further divided into Mongolian, Kazakh, and Tibetan sheep groups, exhibiting distinct geographical distributions and morphological characteristics [46].
In goats, population genetic structure analyses have shown high genetic similarity between Wuxue goats and Guizhou White goats, Chengdu Brown goats, and Jintang black goats, with significant genetic differentiation from other goat breeds [47]. Analysis of 20 Longlin goats and 66 genomes from 7 global breeds revealed that Longlin goats exhibit low linkage disequilibrium and large effective population size [48]. Genomic studies of 30 Guizhou black goats indicated shared ancestry with Shaanbei white cashmere, Yunshang black, Iran indigenous, and Moroccan goats. Guizhou black goats demonstrated significantly higher genetic diversity and lower linkage disequilibrium than other goat breeds [49]. Research on highland and lowland Leizhou goats revealed significant genetic differentiation between the two groups. Genome-wide association studies (GWASs) identified significant associations between genetic variations on NC_030818.1 and leg length traits [50]. Xiao et al. [51] successfully assembled the goat Y chromosome and analyzed resequencing data from 96 global goats. Their findings revealed two major paternal lineages (Y1 and Y2) in pre-domestication goats, a maternal bottleneck effect 2000 years ago, and global dispersal of A haplogroup goats from the Near East. Evidence from Y chromosomes suggested unbalanced gender contributions during goat trade and population expansion after the Neolithic period.

3.1.2. Genetic Structure of Bovine

Recent studies have provided deeper insights into the genetic structure and population differentiation among various cattle breeds. Analysis of resequencing data from 14 Chinese cattle breeds revealed a gradient distribution of mixed Chinese taurine and indicine cattle from northern to southern China [52]. To investigate the genetic composition of Xinjiang brown cattle, genomes from 50 individuals were compared with those of eight global breeds. Results showed that Xinjiang brown cattle inherited 9.88% of their genetic material from Kazakh cattle, contributing environmental adaptation genes, and 90.12% from Swiss brown cattle, providing production-related genes [53]. Bohai black cattle exhibited mixed taurine and indicine ancestry, with close genetic relationships to Jiaxian red and Luxi cattle, and showed high genetic diversity [54]. Genome analysis of 29 Loudi cattle and 96 individuals from global cattle breeds indicated that Loudi cattle originated from East Asian, Chinese, European taurine, and Indian taurine lineages [55]. In order to explore the genetic structural relationship between the Pinan cattle population and other populations, phylogenetic trees, clustering analysis, and population structure analysis were performed. It is more similar to the northern Chinese breeds (Zhangmu cattle, Anxi cattle, Qaidam cattle, Yanbian cattle, and Xinjiang Brown cattle), and significantly different from the southern Chinese breeds (Leiqiong cattle, Wannan cattle, Wenshan cattle, Xiangxi cattle, and Dianzhong cattle) [56].
In yaks, genetic structure analysis of 198 individuals from 6 breeds (Muli, Jinchuan, Changtai, Maiwa, Zhongdian, and Tibetan) revealed that Muli yaks formed a distinct population, while Maiwa yaks exhibited complex genetic structure with gene flow from Jinchuan and Changtai yaks [57]. Molecular marker identification and population structure analysis of Subei yaks indicated they form an independent group within Chinese yak populations [58]. Resequencing data from 91 domestic yaks and 1 wild yak suggested that yak domestication likely originated in the southeastern Qinghai–Tibet Plateau before spreading westward and northeastward, with breed distribution closely linked to historical trade routes and gene flow among domestic, wild, and Tibetan yaks [59]. Single-cell RNA sequencing and resequencing of Tianzhu white yaks revealed significant genetic differentiation from other populations, while gene flow was observed among Pali and Sibu yaks in high-altitude regions [60].
These findings enhance our understanding of the genetic background of cattle breeds and provide valuable guidance for future conservation, genetic resource utilization, population improvement, and adaptive breeding programs.

3.2. Assessment of Genetic Diversity

In recent years, the rapid progression of industrialization and urbanization has significantly impacted natural environments, posing unprecedented challenges to the genetic diversity of many ruminant species. Some rare breeds of sheep, goats, and cattle are facing population declines and even endangerment due to a sharp reduction in genetic diversity [61]. In this context, precise and comprehensive assessments of genetic diversity in ruminants have become urgent. Understanding the distribution of gene frequencies and genetic variation levels can provide a scientific foundation for developing conservation strategies for endangered ruminants [62]. These efforts not only contribute to biodiversity conservation but also offer valuable insights for optimizing existing breeds and developing new breeds in animal husbandry.

3.2.1. Genetic Diversity in Sheep and Goats

Recent studies have analyzed the genetic diversity of 11 Tibetan sheep populations, revealing low genetic diversity, moderate genetic differentiation, and a gradual increase in effective population size over time [63]. Comparative genomic analysis of Chaka sheep (CKA) with other Chinese sheep breeds (Bayinbuluke sheep, Tan sheep, and Oula sheep) was conducted, incorporating the length of runs of homozygosity (ROH) and linkage disequilibrium (LD) decay. The results revealed that the genetic diversity of CKA and TAN was significantly higher than that of OLA and BYK. Furthermore, the inbreeding coefficient (FROH) calculated based on ROH indicated lower inbreeding levels in CKA and Tan, suggesting milder inbreeding depression and relatively better genetic health status [64]. Analysis of 35 Liangshan semi-fine-wool sheep (LSS) using genomic relationship matrices (GRM) indicated low genetic relatedness among most individuals (genetic relatedness based on SNPs < 0.05). The observed heterozygosity (Ho) and expected heterozygosity (He) across all SNP loci were 0.330 and 0.334, respectively, indicating high genetic diversity in the breed [65].
Comparative genomic analyses of Xiangdong black goats and six other Chinese goat breeds showed moderate genetic diversity, low inbreeding, and large effective population sizes in Xiangdong black goats, reflecting a random mating pattern within the population [66]. Genomic analyses of 16 Hainan black goats and 71 individuals from six other geographically distinct goat breeds revealed that the genetic diversity of Hainan black goats was closely linked to their geographic distribution. PCA and phylogenetic tree analysis showed that Hainan black goats were genetically similar to Leizhou goats, consistent with their geographical proximity [67]. Further analysis of SNP density, ROH, iHS, effective population size, and nucleotide diversity in Hainan black goats identified 23,608,983 SNPs. ROH analysis suggested a moderate level of inbreeding in this breed [68]. Genomic resequencing of 123 cashmere goats assessed inbreeding levels, revealing that Inner Mongolian cashmere goats had the lowest inbreeding coefficient (0.0263). A total of 57,224 ROHs and 74 ROH islands were identified [69]. These studies highlight the complex genetic relationships and population histories among different goat breeds, emphasizing the importance of conserving genetic diversity and providing valuable genetic information for future improvement and management of sheep and goat populations.

3.2.2. Genetic Diversity in Bovine

Genetic diversity studies of Yunling cattle revealed low levels of nucleotide diversity [70]. Analysis of Qinchaun cattle showed high genetic diversity, identifying over 20 million SNPs and an IBS distance of 0.243. Some individuals exhibited close genetic relationships. Additionally, 8258 ROHs were detected, and the inbreeding coefficient was calculated as 0.039, indicating moderate inbreeding, especially in bulls [71]. Genome analysis of 20 Kashmir cattle and other breeds showed low inbreeding and high nucleotide diversity, reflecting strong genetic diversity in Kashmir cattle [72]. Genetic diversity assessments of Nanyang cattle revealed closer genetic relationships with central and northern Chinese cattle breeds than with European breeds, higher genetic diversity, and low inbreeding levels. Selection signals related to reproduction and immunity were also identified [73]. A comparative analysis of 23 Xiangxi cattle and 78 genomes from 6 globally representative breeds revealed weak artificial selection pressures and high genomic diversity in Xiangxi cattle, as shown by nucleotide diversity, inbreeding coefficients, linkage disequilibrium decay, and ROH analyses [74]. Comparison of 23 Zaobei cattle with 46 Simmental cattle demonstrated clear genetic separation between the two breeds, confirming the genetic uniqueness of Zaobei cattle [75].
These whole-genome resequencing studies comprehensively evaluated the genetic diversity and inbreeding levels of various cattle breeds, uncovering their unique genetic characteristics and adaptive traits. These findings provide crucial insights for genetic resource conservation and breed improvement in cattle populations.

3.3. Identification of Functional Genes Associated with Economic Traits

In modern animal husbandry, ruminants are among the most important livestock species, and improving their economic traits—such as wool yield, meat quality, reproductive performance, and growth rate—is crucial for enhancing productivity and economic returns [76]. The genetic basis of these traits is directly linked to breed improvement and the sustainable development of the industry. Advances in molecular biology, particularly genomics and bioinformatics, have enabled the identification and understanding of key functional genes underlying these important economic traits [77].

3.3.1. Key Functional Genes in Sheep and Goats

Recent whole-genome resequencing (WGRS) studies on sheep and goats have revealed genomic regions and candidate genes associated with key economic traits, including reproduction, growth, and immune response. By aligning the Hu sheep genome to the reference genome, a total of 39,467,233 SNPs and 8,677,193 insertions/deletions (InDels) were identified. Selection signature analysis revealed genetic selection signals associated with litter size, pinpointing the CC2D1B gene and other reproduction-related genes. Additionally, specific missense mutations and linkage disequilibrium patterns were detected in Hu sheep [78]. In Hu sheep, missense mutations in GPR35 and NAV1 were significantly associated with immunity and growth traits [79]. Growth-related candidate genes such as HDAC1, ACVR1, and GNAI2 in Hu sheep, and RBBP8, PLAT, and CRB1 in Gangba sheep, were identified. Both populations showed strong selection for the CYP2E1 gene, associated with growth [80]. In fine-wool sheep, selective scans in horned and polled groups revealed strong selection on the RXFP2 gene [81]. Wool and cashmere fibers are highly valued for their unique properties and play a significant role in various industries. Studies have shown that wool-related traits exhibit substantial heritability, indicating strong genetic control over these traits and the potential for improvement through selective breeding. This finding provides a critical theoretical foundation for the genetic enhancement and breeding of wool traits [82]. Genome-wide association studies (GWASs) on wool traits in Alpine Merino, Chinese Merino, Qinghai fine-wool sheep, and Aohan fine-wool sheep identified SNPs associated with wool length, fiber diameter, greasy fleece weight, and clean fleece yield, along with estimates of heritability for these traits [83]. Comparative resequencing of wild and domesticated sheep revealed nonsynonymous mutations in candidate genes, with PDGFD emerging as a key gene influencing tail fat deposition in sheep [84]. Researchers conducted resequencing and genome-wide association analysis on Ujimqin sheep with multiple vertebrae and determined that the ABCD4 gene is associated with the number of vertebrae in sheep. The transcriptome expression profile of developing mouse embryos shows that ABCD4 is highly expressed during the critical period of vertebral body formation (4.5–7.5 days). The ABCD4 gene has two missense mutations, and the mutation at position 22 (Chr7: 89393414, C>T) converts arginine to glutamine, which hinders the function of the protein in vertebral development [85].
In dairy and non-dairy goat breeds, WGRS analysis of 89 individuals identified candidate genes related to milk production (GHR, DGAT2), reproduction (PTGS2, ESR2), and immunity (POU2F2, LRRC66) [86]. Genomic analysis of eight local and imported dairy goat breeds revealed key candidate genes for milk production (STK3, PRELID3B), reproduction (ATP5E), growth (CTSZ, GHR), and immune function (CTSZ, NELFCD) [87]. GWAS in Dazu black goats, known for high reproductive performance, identified reproduction-related genes such as ATP1A1 and FGFR2 (udder traits), and GASK1B and ENSCHIG00000026285 (litter size) [88]. Genomic studies of Lanzhou fat-tailed sheep identified genes associated with androgen synthesis and follicle-stimulating hormone response (SRD5A2, SRD5A3, and PAWR) that may contribute to intersex traits. Copy number variations (CNVs) in chromosomes 1, 4, 9, and 16 were also implicated in the expression of genes influencing these traits [89]. Analysis of six multi- and single-kid female goats identified reproductive candidate genes such as AURKA, ENDOG, CDC25C, and NANOS3, which are mainly involved in protein coiled-coil structure and ovarian development [90].
These findings provide valuable molecular markers and candidate genes for genetic improvement and breeding programs in sheep and goats, enhancing our understanding of the mechanisms underlying key economic traits such as reproduction, milk production, and immunity.

3.3.2. Key Functional Genes in Bovine

Elucidating the functions and regulatory mechanisms of genes associated with economic traits in cattle is of significant importance for gaining a deeper understanding of the genetic basis of trait formation and advancing molecular breeding practices in cattle. In Dabieshan cattle, selective scans identified candidate genes associated with reproduction (GPX5, GPX6), feed efficiency (SLC2A5), immune response (IGLL1), heat tolerance (DnaJC1), fat deposition (MLLT10), and coat color (ASIP) [91]. For Xianan cattle, a hybrid of Charolais and Nanyang breeds with robust body size and high meat yield, analysis of 30 individuals and 178 published genomes revealed candidate genes related to skeletal development (NR6A1), meat quality (MCCC1), growth (WSCD1), and immunity (IL11RA, GOLM1) [92]. Annotation of 20 Chaling cattle genomes identified strong selection signals in genes related to immunity, heat tolerance, reproduction, growth, and meat quality [93]. Genomic analysis of Yunling cattle identified 3728 CNV regions, 111 of which overlapped with 76 quantitative trait loci (QTLs) for traits such as subcutaneous fat thickness, longissimus dorsi muscle area, and marbling score [94].
Analysis of whole-genome resequencing data from Hanwoo cattle identified a series of candidate genes that may play critical regulatory roles in milk production, meat quality traits (including tenderness, juiciness, and marbling), and the genetic mechanisms underlying the yellow coat color phenotype [95]. CNV analysis across major water buffalo breeds worldwide mapped CNV regions associated with growth, body weight, and horn development [96]. In yaks, a vital resource on the Qinghai–Tibet Plateau, genomic studies identified 203 selection regions and 27 genes linked to traits such as short stature, milk quality, meat quality, fertility, adaptability, growth, and immunity [97]. Comparative analysis of long-haired and normal-haired Tianzhu white yaks identified 80 genes associated with long-hair traits, including those related to lipid metabolism and cell migration, such as COL22A1, GK5, and SLIT3 [98]. The Jinchuan yak, known for its additional thoracic vertebrae, revealed 330 markers associated with this trait. Validation in 51 yaks confirmed 7 markers with high predictive accuracy. Candidate genes PPP2R2B and TBLR1 were identified for the unique vertebral number [99]. These findings provide a robust scientific basis for future cattle breed improvement and genetic breeding strategies. A summary of candidate genes associated with economic traits in ruminants are presented in Table 1.

3.4. Studies on Adaptive Traits

Understanding the mechanisms underlying the adaptation of cattle and sheep to extreme climates, high altitudes, and various pathogens is crucial for guiding breeding programs, optimizing husbandry practices, and safeguarding the health and welfare of these populations. Research on adaptive traits not only enhances the understanding of the biology of livestock but also provides a scientific basis for addressing challenges posed by climate and environmental changes, supporting global agriculture and environmental management [100].

3.4.1. Adaptive Traits in Sheep and Goats

Recent studies have investigated the genetic adaptation of Chinese indigenous sheep to different climatic conditions, identifying candidate genes associated with adaptation to extreme environments. By aligning to the sheep reference genome and conducting genomic analyses of Kazakh sheep (KAZ), Mongolian sheep (MON), Tibetan sheep (TIB), and Yunnan sheep (YUN), candidate genes associated with adaptation to arid regions (TBXT, TG, HOXA1), high-altitude environments (DYSF, PDGFD, NF1), and warm climates (TSHR, ABCD4, TEX11) were identified [101]. Analysis of copy number variations (CNVs) in 24 Tibetan sheep identified variations in the RUNX1 gene, which is associated with hypoxic adaptation, enabling Tibetan sheep to thrive in the low-oxygen environment of the Qinghai–Tibet Plateau [102]. RUNX1 plays essential roles in biological processes such as bone development [103], immune cell development [104], and hypoxia-related gene expression regulation [105], highlighting its significance in high-altitude adaptation. Further research identified 338 breed-specific selective regions in Chinese indigenous sheep, including CYP17, associated with hypoxic adaptation in Tibetan sheep, and DNAJB5, related to heat tolerance in Duolang sheep [106]. In Hainan black goats, 33 candidate genes were linked to heat tolerance (GNG2, MAPK8) and stress resistance (TLR2, IFI44, ENPP1) [68].

3.4.2. Adaptive Traits in Bovine

Extensive studies have explored the genetic basis of adaptation in cattle to high-altitude and extreme climatic conditions, identifying genetic markers and candidate genes involved in these processes. Genomic analysis of nine Yanbian cattle identified cold tolerance genes such as CORT, FGF5, and CD36 [107]. Comparison of CNVs in Yakut cattle and Holstein cattle with those of Korean and Holstein breeds revealed CNVRs associated with cold adaptation [108]. Genomic analysis of Gansu Meiren yaks identified candidate genes such as BMP2 and ACVR2B, related to high-altitude and hypoxic adaptation [109].
Analysis of genomes from 32 breeds, including 258 cattle, revealed introgression signals between yaks and Tibetan cattle involving genes linked to hypoxia response (EGLN1), cold adaptation (LRP11), DNA damage repair (LATS1), and UV radiation resistance (GNPAT). SNPs in the EGLN1 promoter region reduced its expression, enhancing hypoxia tolerance in Tibetan cattle [110]. SNP functional annotation in Siberian cattle identified chromosomal regions and candidate SNPs associated with thermoregulation and genes linked to heat adaptation, such as GRIA4 and COX17 [111]. Genomic analysis of Lincang humped cattle, which contain indicine and taurine ancestry, identified a missense mutation in HELB related to heat tolerance, providing insights into indicine cattle’s adaptation to hot environments [112].
CNV and CNVR analyses in Chinese, Indian, and European commercial cattle breeds identified genes associated with environmental adaptation (CNGB3, FAM161A, ERC2) and oxidative stress and anti-inflammatory responses (COMMD1, OXR1) [113]. Comparative genomic analysis of Xinjiang Mongolian cattle and 134 genomes from 9 representative breeds worldwide revealed candidate genes related to desert adaptation, including genes involved in water reabsorption, osmotic regulation, metabolism, and energy balance [114]. Genotyping and resequencing of Chinese indigenous cattle genomes revealed candidate genes related to thermogenesis and energy metabolism, such as UQCR11, EGR1, and STING1 [115]. Studies of Iran indigenous river buffalo populations identified a region on chromosome 2 overlapping the OCA2-HERC2 gene, indicating a genetic basis for UV adaptation and pigmentation mechanisms in water buffalo [116]. Genomic analysis of 25 domesticated buffalo breeds revealed selection signals in swamp buffalo related to neural system genes and in river buffalo associated with heat stress and immune genes [117]. Comparison of salt-tolerant Chilika buffalo and non-salt-tolerant Murrah buffalo identified pathways related to salt tolerance, including MAPK signaling and renin secretion [118]. Genomic analysis of Xiangxi white buffalo (XWB) identified candidate genes associated with nervous system traits (GRIK2), reproduction (KCNIP4), growth and development (IFNAR1), morphology (LINGO2), and immunity (IRAK3) [119].
These findings deepen the understanding of the genetic basis of adaptation in cattle breeds and provide critical scientific tools for future genetic improvement and breeding programs focused on environmental adaptability. A summary of candidate genes associated with adaptive traits in ruminants can be found in Table 2.

4. Conservation of Genetic Resources

Ruminants, a vital component of global agriculture, play a crucial role in ensuring food security, maintaining ecological balance, and supporting rural livelihoods. They provide us with essential products such as meat, milk, and hides, while also contributing to soil fertility and nutrient cycling [120]. However, the genetic resources of these invaluable animals are under increasing threat. The widespread adoption of a few high-yielding breeds has led to the erosion of genetic diversity, leaving ruminant populations vulnerable to diseases, environmental changes, and market fluctuations. The loss of indigenous breeds, each uniquely adapted to specific environments and production systems, further exacerbates this problem [121]. The conservation of ruminant genetic resources is thus an urgent task. In the face of such a challenging situation, the whole-genome resequencing (WGRS) technology has emerged as a ray of hope.
WGRS technology has played a pivotal role not only in advancing the study of genetic diversity and gene function but also in the development, conservation, and utilization of genetic resources [122]. This technology enables detailed analysis of species’ genetic diversity and provides a powerful tool for monitoring and protecting biodiversity. By examining genetic structures and population histories, WGRS facilitates the elucidation of species’ evolutionary processes, offering scientific insights for the conservation and management of endangered species.

4.1. Conservation of Genetic Resources in Sheep and Goats

The Inner Mongolian Cashmere goat (IMCG), a premium cashmere breed in China, has been the focus of conservation efforts. To effectively preserve and utilize the purebred genetic resources of IMCG, researchers performed WGRS on 225 randomly selected individuals. The results showed low levels of runs of homozygosity (ROH), a low mean inbreeding coefficient, and high genetic diversity within the IMCG population [123]. In Tibetan sheep, WGRS analyses of 11 populations indicated low genetic diversity, minimal genetic differentiation, and clear population structures. Effective population sizes increased over time, and evolutionary relationships among populations were closely associated with geographical and genetic distances, reflecting the profound influence of historical activities on Tibetan sheep populations [124]. Genomic data analysis of Baiyu black goats and Chuanzhong black goats was conducted to assess genetic diversity, population structure, and selection signatures. The results revealed that Baiyu black goats exhibit greater genetic diversity and significant genetic differentiation compared to Chuanzhong black goats. Phylogenetic analysis indicated that Baiyu black goats are more closely related to Tibetan cashmere goats, while Chuanzhong black goats show closer genetic affinity to Chengdu gray goats. These findings provide a theoretical foundation for the further conservation of genetic resources in Baiyu black goats and Chuanzhong black goats [125].

4.2. Conservation of Genetic Resources in Bovine

Kazakh cattle, known for their adaptability and versatility in Xinjiang, were analyzed to study their evolutionary history, breeding characteristics, and germplasm resource conservation. Researchers compared genomes from 26 Kazakh cattle with 103 genomes from 7 other global cattle breeds. The findings revealed large effective population sizes, low linkage disequilibrium, and a population structure predominantly derived from European taurine ancestry [126]. Kongshan cattle, recognized for their resilience in harsh environments, have experienced population declines due to competition with exotic breeds. Using WGRS, researchers identified substantial genetic variation in this breed, including mutations in NEIL2 and PNKP, which are associated with stress resistance. These findings are instrumental in conserving the genetic diversity of Kongshan cattle [127]. To conserve and utilize the endangered Dengchuan cattle, a unique breed in Yunnan Province, researchers sequenced 10 individuals. The results revealed a high level of hybridization, with positive selection signals associated with milk production, disease resistance, growth, and heat tolerance. These findings provide a theoretical basis for understanding the genetic mechanisms underlying Dengchuan cattle’s desirable traits, such as high milk yield, adaptability, roughage tolerance, immune performance, and small body size [128].
In conclusion, the application of WGRS has not only deepened the understanding of genetic diversity and population structures in cattle and sheep but has also provided a robust tool for the effective conservation and sustainable utilization of genetic resources. This technology holds significant scientific value for maintaining biodiversity and promoting sustainable agricultural development.

5. Deciphering Complex Traits in Ruminants Using Whole-Genome Resequencing: Challenges and Multi-Omics Breakthroughs

Whole-genome resequencing (WGRS) has played a significant role in the analysis of genetic diversity, genomic selection [129], disease research [130], and genetic resource conservation in ruminants, providing a powerful tool for ruminant genetics and breeding research. However, despite its notable advantages in many aspects, WGRS still faces certain limitations in deciphering complex traits.
Complex traits are influenced by multiple genes and environmental factors, and their inheritance patterns do not conform to classical Mendelian genetics [131]. Unlike simple traits controlled by single genes, complex traits typically exhibit quantitative characteristics, such as milk yield [132], growth rate [133], and wool traits [134] in ruminants. The genetic basis of these traits involves multiple genes, each with small effects, and may include gene–gene interactions (epistasis) and gene–environment interactions. Furthermore, the phenotypic variation of complex traits is often continuous, making it difficult to interpret through simple genetic models [135]. Although WGRS can comprehensively detect genomic variations, it still encounters challenges in unraveling the polygenic regulatory networks, gene interactions, and environmental influences underlying complex traits.
To fully elucidate the genetic mechanisms of complex traits, WGRS needs to be integrated with multiple technologies to construct a multi-level research framework. Transcriptome sequencing (RNA-seq) serves as a crucial complementary technology, linking genomic variants identified by WGRS to functional gene expression changes and revealing the impact of variants on gene regulation [136]. Epigenomic technologies, such as DNA methylation sequencing and chromatin accessibility analysis, can further dissect the role of epigenetic modifications in complex traits, elucidating how environmental factors influence gene expression through epigenetic regulation [137]. Additionally, metabolomics and proteomics provide insights into downstream metabolic products and protein levels, helping to construct a comprehensive regulatory network from genomic variation to phenotypic manifestation [138]. In terms of data analysis, integrating systems biology approaches and machine learning algorithms with multi-omics data enables a more comprehensive understanding of the contributions of gene–gene and gene–environment interactions to complex traits [139].
By combining WGRS with transcriptomics, epigenomics, metabolomics, proteomics, and other multi-omics technologies, and complementing these with systems biology analysis and functional validation experiments, the genetic basis of complex traits can be fully deciphered. This integrated approach provides a scientific foundation for genetic improvement and precision breeding in ruminants.

6. Future Perspectives

With the continuous advancement of high-throughput sequencing technologies and declining sequencing costs, WGRS has gained widespread application. Given the massive datasets generated by resequencing, future research will increasingly depend on the development of advanced bioinformatics tools and algorithms to efficiently and accurately interpret these data, enhancing the efficiency and precision of data analysis [140]. Improvements in computing power, memory, storage technologies, and data processing capabilities have driven the adoption of machine learning algorithms in biomedical research [141], and these algorithms also show significant potential in WGRS studies of ruminants. For instance, clustering algorithms can efficiently classify and organize resequencing data, rapidly identifying characteristic gene sequences in different ruminant populations, thus providing more efficient methods for analyzing population genetic structures [142]. Deep learning algorithms, by learning from large datasets of known functional gene–trait associations, can establish precise predictive models. These models have the potential to accurately predict the functions and roles of unknown genes in economic and adaptive traits, significantly reducing research timelines [143].
While WGRS focuses on analyzing genetic differences at the genomic level, multi-omics (integrating genomics, epigenomics, transcriptomics, proteomics, and metabolomics) provides a systematic approach to understanding complex biological phenomena and mechanisms by combining multiple layers of data [144]. For example, common variable immunodeficiency (CVID), caused by multigenic variations and characterized by diverse clinical manifestations, can be better understood through multi-omics analysis, revealing the underlying molecular complexity of the disease [145]. The integration of WGRS and multi-omics analysis enables efficient identification of key genetic information and systematic elucidation of the molecular regulatory networks and genetic mechanisms underlying complex traits, thereby advancing livestock breeding programs. With continuous theoretical advancements and practical innovations, significant breakthroughs in this field are anticipated in the near future.

7. Conclusions

This review provides a concise overview of the principles and advantages of whole-genome resequencing (WGRS) technology and systematically explores its applications and recent advancements in ruminant research. In the section on “Applications and Research Progress in Ruminants,” it highlights the use of WGRS for analyzing population genetic structures, elucidating the genetic relationships and evolutionary history among different ruminant populations. Through the assessment of genetic diversity, it further clarifies the genomic diversity characteristics and genetic backgrounds of various ruminant breeds. Additionally, the review delves into the application of WGRS in identifying functional genes associated with economic traits (such as meat quality and milk yield) and adaptive traits, emphasizing its critical role in enhancing livestock production efficiency and breeding precision. However, despite the significant advantages of WGRS in detecting genomic variations, it still faces challenges and limitations in deciphering the polygenic regulatory networks and gene–environment interactions underlying complex traits.

Author Contributions

Conceptualization, Q.W., Y.L., M.L., Z.G., J.W. and W.D.; writing—original draft preparation, Q.W.; investigation, Y.L., M.L. and Z.G.; writing—review and editing, Y.L., M.L. and Z.G.; visualization, Y.L. and Z.G.; writing—assessment and refinement, D.L., Y.G., J.W. and W.D.; supervision, D.L., Y.G., J.W. and W.D.; project administration, J.W.; funding acquisition, W.D. and J.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Nature Science Foundation of China (32160771, 32302707), Yunnan Provincial Agricultural Union Foundation (202101BD070001-006), Major Science and Technology Projects in Yunnan Province (202202AE090005), First level project of the National Key Research and Development Program (2022YFD1100408), and Yunnan Province Agricultural Joint Special Project—Key Project (202101BD070001-006).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bovine Genome Sequencing and Analysis Consortium; Elsik, C.G.; Tellam, R.L.; Worley, K.C.; Gibbs, R.A.; Muzny, D.M.; Weinstock, G.M.; Adelson, D.L.; Eichler, E.E.; Elnitski, L.; et al. The genome sequence of taurine cattle: A window to ruminant biology and evolution. Science 2009, 324, 522–528. [Google Scholar] [PubMed]
  2. Qiu, Q.; Zhang, G.; Ma, T.; Qian, W.; Wang, J.; Ye, Z.; Cao, C.; Hu, Q.; Kim, J.; Larkin, D.M.; et al. The yak genome and adaptation to life at high altitude. Nat. Genet. 2012, 44, 946–949. [Google Scholar] [CrossRef] [PubMed]
  3. Yang, Y.; Wang, Y.; Zhao, Y.; Zhang, X.; Li, R.; Chen, L.; Zhang, G.; Jiang, Y.; Qiu, Q.; Wang, W.; et al. Draft genome of the Marco Polo Sheep (Ovis ammon polii). Gigascience 2017, 6, gix106. [Google Scholar] [CrossRef] [PubMed]
  4. Low, W.Y.; Tearle, R.; Bickhart, D.M.; Rosen, B.D.; Kingan, S.B.; Swale, T.; Thibaud-Nissen, F.; Murphy, T.D.; Young, R.; Lefevre, L.; et al. Chromosome-level assembly of the water buffalo genome surpasses human and goat genomes in sequence contiguity. Nat. Commun. 2019, 10, 260. [Google Scholar] [CrossRef]
  5. Wu, H.; Luo, L.Y.; Zhang, Y.H.; Zhang, C.Y.; Huang, J.H.; Mo, D.X.; Zhao, L.M.; Wang, Z.X.; Wang, Y.C.; He-Hua, E.; et al. Telomere-to-telomere genome assembly of a male goat reveals variants associated with cashmere traits. Nat. Commun. 2024, 15, 10041. [Google Scholar] [CrossRef]
  6. Bentley, D.R. Whole-Genome Re-Sequencing. Curr. Opin. Genet. Dev. 2006, 16, 545–552. [Google Scholar] [CrossRef]
  7. Lai, Y.-T.; Yeung, C.K.L.; Omland, K.E.; Pang, E.-L.; Hao, Y.; Liao, B.-Y.; Cao, H.-F.; Zhang, B.-W.; Yeh, C.-F.; Hung, C.-M.; et al. Standing Genetic Variation as the Predominant Source for Adaptation of a Songbird. Proc. Natl. Acad. Sci. USA 2019, 116, 2152–2157. [Google Scholar] [CrossRef]
  8. Liang, Z.; Duan, S.; Sheng, J.; Zhu, S.; Ni, X.; Shao, J.; Liu, C.; Nick, P.; Du, F.; Fan, P.; et al. Whole-Genome Resequencing of 472 Vitis Accessions for Grapevine Diversity and Demographic History Analyses. Nat. Commun. 2019, 10, 1190. [Google Scholar] [CrossRef]
  9. Li, Y.; Cao, K.; Zhu, G.; Fang, W.; Chen, C.; Wang, X.; Zhao, P.; Guo, J.; Ding, T.; Guan, L.; et al. Genomic Analyses of an Extensive Collection of Wild and Cultivated Accessions Provide New Insights into Peach Breeding History. Genome Biol. 2019, 20, 36. [Google Scholar] [CrossRef]
  10. Athanasopoulou, K.; Boti, M.A.; Adamopoulos, P.G.; Skourou, P.C.; Scorilas, A. Third-Generation Sequencing: The Spearhead towards the Radical Transformation of Modern Genomics. Life 2021, 12, 30. [Google Scholar] [CrossRef]
  11. Olivucci, G.; Iovino, E.; Innella, G.; Turchetti, D.; Pippucci, T.; Magini, P. Long read sequencing on its way to the routine diagnostics of genetic diseases. Front. Genet. 2024, 15, 1374860. [Google Scholar] [CrossRef] [PubMed]
  12. Ma, L.; Yang, C.; Xiao, D.; Liu, X.; Jiang, X.; Lin, H.; Ying, Z.; Lin, Y. Chromosome-level assembly of Dictyophora rubrovolvata genome using third-generation DNA sequencing and Hi-C analysis. G3 2023, 13, jkad102. [Google Scholar] [CrossRef]
  13. Satam, H.; Joshi, K.; Mangrolia, U.; Waghoo, S.; Zaidi, G.; Rawool, S.; Thakare, R.P.; Banday, S.; Mishra, A.K.; Das, G.; et al. Next-Generation Sequencing Technology: Current Trends and Advancements. Biology 2023, 12, 997. [Google Scholar] [CrossRef] [PubMed]
  14. Xu, F.; Chen, H.; Chen, C.; Liu, J.; Song, Z.; Ding, C. The Mutagenic Effect of Cold Plasma on Medicago sativa L. Free Radic. Biol. Med. 2024, 223, 18–29. [Google Scholar] [CrossRef]
  15. Fuentes-Pardo, A.P.; Ruzzante, D.E. Whole-Genome Sequencing Approaches for Conservation Biology: Advantages, Limitations and Practical Recommendations. Mol. Ecol. 2017, 26, 5369–5406. [Google Scholar] [CrossRef]
  16. Maybery-Reupert, K.; Isenegger, D.; Hayden, M.; Cogan, N. Development of Genomic Resources for Rhodes Grass (Chloris gayana), Draft Genome and Annotated Variant Discovery. Front. Plant Sci. 2023, 14, 1239290. [Google Scholar] [CrossRef]
  17. Guo, F.; Ye, Y.; Zhu, K.; Lin, S.; Wang, Y.; Dong, Z.; Yao, R.; Li, H.; Wang, W.; Liao, Z.; et al. Genetic Diversity, Population Structure, and Environmental Adaptation Signatures of Chinese Coastal Hard-Shell Mussel Mytilus coruscus Revealed by Whole-Genome Sequencing. Int. J. Mol. Sci. 2023, 24, 13641. [Google Scholar] [CrossRef]
  18. Bhattarai, G.; Shi, A.; Mou, B.; Correll, J.C. Resequencing Worldwide Spinach Germplasm for Identification of Field Resistance QTLs to Downy Mildew and Assessment of Genomic Selection Methods. Hortic. Res. 2022, 9, uhac205. [Google Scholar] [CrossRef]
  19. Zhang, W.; Lu, Z.; Guo, T.; Yuan, C.; Liu, J. Construction of a High-Density Genetic Map and QTL Localization of Body Weight and Wool Production Related Traits in Alpine Merino Sheep Based on WGR. BMC Genom. 2024, 25, 641. [Google Scholar] [CrossRef]
  20. Feng, Y.; Yang, C.; Zhang, J.; Qiao, J.; Wang, B.; Zhao, Y. Construction of a High-Density Paulownia Genetic Map and QTL Mapping of Important Phenotypic Traits Based on Genome Assembly and Whole-Genome Resequencing. Int. J. Mol. Sci. 2023, 24, 15647. [Google Scholar] [CrossRef]
  21. Ma, Y.; Jia, J.; Fan, R.; Lu, Y.; Zhao, X.; Zhong, Y.; Yang, J.; Ma, L.; Wang, Y.; Lv, M.; et al. Screening and Identification of the First Non-CRISPR/Cas9-Treated Chinese Miniature Pig with Defective Porcine Endogenous Retrovirus Pol Genes. Front. Immunol. 2021, 12, 797608. [Google Scholar] [CrossRef] [PubMed]
  22. Li, P.; Tan, X.; Liu, R.; Rahman, F.U.; Jiang, J.; Sun, L.; Fan, X.; Liu, J.; Liu, C.; Zhang, Y. QTL Detection and Candidate Gene Analysis of Grape White Rot Resistance by Interspecific Grape (Vitis vinifera L. × Vitis davidii Foex.) Crossing. Hortic. Res. 2023, 10, uhad063. [Google Scholar] [CrossRef] [PubMed]
  23. Bourgeois, Y.X.C.; Warren, B.H. An Overview of Current Population Genomics Methods for the Analysis of Whole-Genome Resequencing Data in Eukaryotes. Mol. Ecol. 2021, 30, 6036–6071. [Google Scholar] [CrossRef] [PubMed]
  24. Kishikawa, T.; Momozawa, Y.; Ozeki, T.; Mushiroda, T.; Inohara, H.; Kamatani, Y.; Kubo, M.; Okada, Y. Empirical Evaluation of Variant Calling Accuracy Using Ultra-Deep Whole-Genome Sequencing Data. Sci. Rep. 2019, 9, 1784. [Google Scholar] [CrossRef]
  25. Jiang, Y.; Jiang, Y.; Wang, S.; Zhang, Q.; Ding, X. Optimal Sequencing Depth Design for Whole Genome Re-Sequencing in Pigs. BMC Bioinform. 2019, 20, 556. [Google Scholar] [CrossRef]
  26. Logsdon, G.A.; Vollger, M.R.; Eichler, E.E. Long-Read Human Genome Sequencing and Its Applications. Nat. Rev. Genet. 2020, 21, 597–614. [Google Scholar] [CrossRef]
  27. Smith, S.D.; Kawash, J.K.; Grigoriev, A. Lightning-Fast Genome Variant Detection with GROM. GigaScience 2017, 6, gix091. [Google Scholar] [CrossRef]
  28. Kosugi, S.; Momozawa, Y.; Liu, X.; Terao, C.; Kubo, M.; Kamatani, Y. Comprehensive Evaluation of Structural Variation Detection Algorithms for Whole Genome Sequencing. Genome Biol. 2019, 20, 117. [Google Scholar] [CrossRef]
  29. Wratten, L.; Wilm, A.; Göke, J. Reproducible, Scalable, and Shareable Analysis Pipelines with Bioinformatics Workflow Managers. Nat. Methods 2021, 18, 1161–1168. [Google Scholar] [CrossRef]
  30. Shin, S.C.; Ahn, D.H.; Kim, S.J.; Lee, H.; Oh, T.-J.; Lee, J.E.; Park, H. Advantages of Single-Molecule Real-Time Sequencing in High-GC Content Genomes. PLoS ONE 2013, 8, e68824. [Google Scholar] [CrossRef]
  31. Levy, S.E.; Boone, B.E. Next-Generation Sequencing Strategies. Cold Spring Harb. Perspect. Med. 2019, 9, a025791. [Google Scholar] [CrossRef] [PubMed]
  32. Basapathi Raghavendra, J.; Zorzano, M.-P.; Kumaresan, D.; Martin-Torres, J. DNA Sequencing at the Picogram Level to Investigate Life on Mars and Earth. Sci. Rep. 2023, 13, 15277. [Google Scholar] [CrossRef] [PubMed]
  33. Hossein-Zadeh, N.G. An overview of recent technological developments in bovine genomics. Vet. Anim. Sci. 2024, 25, 100382. [Google Scholar] [CrossRef] [PubMed]
  34. Didion, J.P.; Yang, H.; Sheppard, K.; Fu, C.-P.; McMillan, L.; de Villena, F.P.-M.; Churchill, G. Discovery of novel variants in genotyping arrays improves genotype retention and reduces ascertainment bias. BMC Genom. 2012, 13, 34. [Google Scholar] [CrossRef]
  35. Saurabh, R.; Fouodo, C.J.K.; König, I.R.; Busch, H.; Wohlers, I. A survey of genome-wide association studies, polygenic scores and UK Biobank highlights resources for autoimmune disease genetics. Front. Immunol. 2022, 13, 972107. [Google Scholar] [CrossRef]
  36. Thanh Nguyen, D.; Hoang Nguyen, Q.; Thuy Duong, N.; Vo, N.S. LmTag: Functional-enrichment and imputation-aware tag SNP selection for population-specific genotyping arrays. Brief. Bioinform. 2022, 23, bbac252. [Google Scholar] [CrossRef]
  37. Runheim, H.; Pettersson, M.; Hammarsjö, A.; Nordgren, A.; Henriksson, M.; Lindstrand, A.; Levin, L.; Soller, M.J. The cost-effectiveness of whole genome sequencing in neurodevelopmental disorders. Sci. Rep. 2023, 13, 6904. [Google Scholar] [CrossRef]
  38. Zhang, S.; Li, J.; Qin, Q.; Liu, W.; Bian, C.; Yi, Y.; Wang, M.; Zhong, L.; You, X.; Tang, S.; et al. Whole-Genome Sequencing of Chinese Yellow Catfish Provides a Valuable Genetic Resource for High-Throughput Identification of Toxin Genes. Toxins 2018, 10, 488. [Google Scholar] [CrossRef]
  39. Jung, H.; Winefield, C.; Bombarely, A.; Prentis, P.; Waterhouse, P. Tools and Strategies for Long-Read Sequencing and De Novo Assembly of Plant Genomes. Trends Plant Sci. 2019, 24, 700–724. [Google Scholar] [CrossRef]
  40. Yang, J.; Cai, Z.; Fang, Y.; Shan, B.; Zhang, R.; Lin, L.; Li, Y.; Zhang, J. Whole-Genome Resequencing Reveals Signatures of Adaptive Evolution in Acanthopagrus latus and Rhabdosargus sarba. Animals 2024, 14, 2339. [Google Scholar] [CrossRef]
  41. Feng, P.; Zeng, T.; Yang, H.; Chen, G.; Du, J.; Chen, L.; Shen, J.; Tao, Z.; Wang, P.; Yang, L.; et al. Whole-genome resequencing provides insights into the population structure and domestication signatures of ducks in eastern China. BMC Genom. 2021, 22, 401. [Google Scholar] [CrossRef] [PubMed]
  42. Li, X.; Han, B.; Liu, D.; Wang, S.; Wang, L.; Pei, Q.; Zhang, Z.; Zhao, J.; Huang, B.; Zhang, F.; et al. Whole-Genome Resequencing to Investigate the Genetic Diversity and Mechanisms of Plateau Adaptation in Tibetan Sheep. J. Anim. Sci. Biotechnol. 2024, 15, 164. [Google Scholar] [CrossRef] [PubMed]
  43. Chong, Y.; Xiong, H.; Gao, Z.; Lu, Y.; Hong, J.; Wu, J.; He, X.; Xi, D.; Tu, X.; Deng, W. Genomic and Transcriptomic Landscape to Decipher the Genetic Basis of Hyperpigmentation in Lanping Black-Boned Sheep (Ovis aries). BMC Genom. 2024, 25, 845. [Google Scholar] [CrossRef]
  44. Zhang, W.; Jin, M.; Li, T.; Lu, Z.; Wang, H.; Yuan, Z.; Wei, C. Whole-Genome Resequencing Reveals Selection Signal Related to Sheep Wool Fineness. Animals 2023, 13, 2944. [Google Scholar] [CrossRef]
  45. Zhang, W.; Jin, M.; Lu, Z.; Li, T.; Wang, H.; Yuan, Z.; Wei, C. Whole Genome Resequencing Reveals Selection Signals Related to Wool Color in Sheep. Animals 2023, 13, 3265. [Google Scholar] [CrossRef]
  46. Zhang, D.-Y.; Zhang, X.-X.; Li, F.-D.; Yuan, L.-F.; Li, X.-L.; Zhang, Y.-K.; Zhao, Y.; Zhao, L.-M.; Wang, J.-H.; Xu, D.; et al. Whole-Genome Resequencing Reveals Molecular Imprints of Anthropogenic and Natural Selection in Wild and Domesticated Sheep. Zool. Res. 2022, 43, 695–705. [Google Scholar] [CrossRef]
  47. Li, C.; Wang, X.; Li, H.; Ahmed, Z.; Luo, Y.; Qin, M.; Yang, Q.; Long, Z.; Lei, C.; Yi, K. Whole-Genome Resequencing Reveals Diversity and Selective Signals in the Wuxue Goat. Anim. Genet. 2024, 55, 575–587. [Google Scholar] [CrossRef]
  48. Chen, Q.; Huang, Y.; Wang, Z.; Teng, S.; Hanif, Q.; Lei, C.; Sun, J. Whole-Genome Resequencing Reveals Diversity and Selective Signals in Longlin Goat. Gene 2021, 771, 145371. [Google Scholar] [CrossRef]
  49. Chang, L.; Zheng, Y.; Li, S.; Niu, X.; Huang, S.; Long, Q.; Ran, X.; Wang, J. Identification of Genomic Characteristics and Selective Signals in Guizhou Black Goat. BMC Genom. 2024, 25, 164. [Google Scholar] [CrossRef]
  50. Liu, J.; Dong, S.; Lv, J.; Li, Y.; Sun, B.; Guo, Y.; Deng, M.; Liu, D.; Liu, G. Screening of SNP Loci Related to Leg Length Trait in Leizhou Goats Based on Whole-Genome Resequencing. Int. J. Mol. Sci. 2024, 25, 12450. [Google Scholar] [CrossRef]
  51. Xiao, C.; Li, J.; Xie, T.; Chen, J.; Zhang, S.; Elaksher, S.H.; Jiang, F.; Jiang, Y.; Zhang, L.; Zhang, W.; et al. The Assembly of Caprine Y Chromosome Sequence Reveals a Unique Paternal Phylogenetic Pattern and Improves Our Understanding of the Origin of Domestic Goat. Ecol. Evol. 2021, 11, 7779–7795. [Google Scholar] [CrossRef] [PubMed]
  52. Naji, M.M.; Jiang, Y.; Utsunomiya, Y.T.; Rosen, B.D.; Sölkner, J.; Wang, C.; Jiang, L.; Zhang, Q.; Zhang, Y.; Ding, X.; et al. Favored Single Nucleotide Variants Identified Using Whole Genome Re-Sequencing of Austrian and Chinese Cattle Breeds. Front. Genet. 2022, 13, 974787. [Google Scholar] [CrossRef]
  53. Chen, Q.; Xu, L.; Zhang, M.; Zhang, T.; Yan, M.; Zhai, M.; Huang, X. Whole Genome Resequencing Reveals the Genetic Contribution of Kazakh and Swiss Brown Cattle to a Population of Xinjiang Brown Cattle. Gene 2022, 839, 146725. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, Z.; Sun, H.; Lai, W.; Hu, M.; Zhang, Y.; Bai, C.; Liu, J.; Ren, H.; Li, F.; Yan, S. Genome-Wide Re-Sequencing Reveals Population Structure and Genetic Diversity of Bohai Black Cattle. Anim. Genet. 2022, 53, 133–136. [Google Scholar] [CrossRef] [PubMed]
  55. Jin, L.; Zhang, B.; Luo, J.; Li, J.; Liang, J.; Wu, W.; Xie, Y.; Li, F.; Lei, C.; Yi, K. Genomics, Origin and Selection Signals of Loudi Cattle in Central Hunan. Biology 2022, 11, 1775. [Google Scholar] [CrossRef]
  56. Bo, D.; Feng, Y.; Bai, Y.; Li, J.; Wang, Y.; You, Z.; Shen, J.; Bai, Y. Whole-Genome Resequencing Reveals Genetic Diversity and Growth Trait-Related Genes in Pinan Cattle. Animals 2024, 14, 2163. [Google Scholar] [CrossRef]
  57. Zhang, S.; Li, J.; Zhao, Y.; Tang, Y.; Li, H.; Song, T.; An, T.; Guan, J.; Li, X.; Zhang, M. Whole-Genome Resequencing Reveals Genetic Diversity, Differentiation, and Selection Signatures of Yak Breeds/Populations in Southwestern China. Front. Genet. 2024, 15, 1382128. [Google Scholar] [CrossRef]
  58. Guo, S.; Yu, T.; Wang, X.; Zhao, S.; Zhao, E.; Ainierlitu; Ba, T.; Gan, M.; Dong, C.; Naerlima; et al. Whole-Genome Resequencing Reveals the Uniqueness of Subei Yak. J. Anim. Sci. 2024, 102, skae152. [Google Scholar] [CrossRef]
  59. Chai, Z.; Xin, J.; Zhang, C.; Dawayangla; Luosang; Zhang, Q.; Pingcuozhandui; Li, C.; Zhu, Y.; Cao, H.W.; et al. Whole-Genome Resequencing Provides Insights into the Evolution and Divergence of the Native Domestic Yaks of the Qinghai-Tibet Plateau. BMC Evol. Biol. 2020, 20, 137. [Google Scholar] [CrossRef]
  60. Zheng, Q.; Wu, X.; Ma, X.; Zhou, X.; Wang, T.; Ma, C.; Zhang, M.; Chu, M.; Guo, X.; Liang, C.; et al. Genetic Structure Analysis of Yak Breeds and Their Response to Adaptive Evolution. Genomics 2024, 116, 110933. [Google Scholar] [CrossRef]
  61. Kawęcka, A.; Pasternak, M.; Miksza-Cybulska, A.; Puchała, M. Native Sheep Breeds in Poland-Importance and Outcomes of Genetic Resources Protection Programmes. Animals 2022, 12, 1510. [Google Scholar] [CrossRef] [PubMed]
  62. Lan, D.; Xiong, X.; Mipam, T.D.; Fu, C.; Li, Q.; Ai, Y.; Hou, D.; Chai, Z.; Zhong, J.; Li, J. Genetic Diversity, Molecular Phylogeny, and Selection Evidence of Jinchuan Yak Revealed by Whole-Genome Resequencing. G3 Genes Genomes Genet. 2018, 8, 945–952. [Google Scholar] [CrossRef] [PubMed]
  63. Tian, D.; Han, B.; Li, X.; Liu, D.; Zhou, B.; Zhao, C.; Zhang, N.; Wang, L.; Pei, Q.; Zhao, K. Genetic Diversity and Selection of Tibetan Sheep Breeds Revealed by Whole-Genome Resequencing. Anim. Biosci. 2023, 36, 991–1002. [Google Scholar] [CrossRef] [PubMed]
  64. Cheng, J.; Zhao, H.; Chen, N.; Cao, X.; Hanif, Q.; Pi, L.; Hu, L.; Chaogetu, B.; Huang, Y.; Lan, X.; et al. Population structure, genetic diversity, and selective signature of Chaka sheep revealed by whole genome sequencing. BMC Genom. 2020, 21, 520. [Google Scholar] [CrossRef]
  65. Sun, X.; Guo, J.; Li, R.; Zhang, H.; Zhang, Y.; Liu, G.E.; Emu, Q.; Zhang, H. Whole-Genome Resequencing Reveals Genetic Diversity and Wool Trait-Related Genes in Liangshan Semi-Fine-Wool Sheep. Animals 2024, 14, 444. [Google Scholar] [CrossRef]
  66. Liu, Z.; Li, H.; Luo, Y.; Li, J.; Sun, A.; Ahmed, Z.; Zhang, B.; Lei, C.; Yi, K. Comprehensive Whole-Genome Resequencing Unveils Genetic Diversity and Selective Signatures of the Xiangdong Black Goat. Front. Genet. 2024, 15, 1326828. [Google Scholar] [CrossRef]
  67. Chen, Q.; Chai, Y.; Zhang, W.; Cheng, Y.; Zhang, Z.; An, Q.; Chen, S.; Man, C.; Du, L.; Zhang, W.; et al. Whole-Genome Sequencing Reveals the Genomic Characteristics and Selection Signatures of Hainan Black Goat. Genes 2022, 13, 1539. [Google Scholar] [CrossRef]
  68. An, Z.X.; Shi, L.G.; Hou, G.Y.; Zhou, H.L.; Xun, W.J. Genetic Diversity and Selection Signatures in Hainan Black Goats Revealed by Whole-Genome Sequencing Data. Anim. Int. J. Anim. Biosci. 2024, 18, 101147. [Google Scholar] [CrossRef]
  69. Zhao, Q.; Huang, C.; Chen, Q.; Su, Y.; Zhang, Y.; Wang, R.; Su, R.; Xu, H.; Liu, S.; Ma, Y.; et al. Genomic Inbreeding and Runs of Homozygosity Analysis of Cashmere Goat. Animals 2024, 14, 1246. [Google Scholar] [CrossRef]
  70. Chen, Q.; Zhan, J.; Shen, J.; Qu, K.; Hanif, Q.; Liu, J.; Zhang, J.; Chen, N.; Chen, H.; Huang, B.; et al. Whole-Genome Resequencing Reveals Diversity, Global and Local Ancestry Proportions in Yunling Cattle. J. Anim. Breed. Genet. 2020, 137, 641–650. [Google Scholar] [CrossRef]
  71. Ma, J.; Fan, A.-P.; Wang, W.-S.; Zhang, J.-C.; Jiang, X.-J.; Ma, R.-J.; Jia, S.-Q.; Liu, F.; Lei, C.-C.; Huang, Y.-Z. Analysis of Genetic Diversity and Genetic Structure of Qinchuan Cattle Conservation Population Using Whole-Genome Resequencing. Yi Chuan Hered. 2023, 45, 602–616. [Google Scholar]
  72. Ahmed, Z.; Xiang, W.; Wang, F.; Nawaz, M.; Kuthu, Z.H.; Lei, C.; Xu, D. Whole-Genome Resequencing Deciphers Patterns of Genetic Diversity, Phylogeny, and Evolutionary Dynamics in Kashmir Cattle. Anim. Genet. 2024, 55, 511–526. [Google Scholar] [CrossRef]
  73. Zhang, Y.; Wei, Z.; Zhang, M.; Wang, S.; Gao, T.; Huang, H.; Zhang, T.; Cai, H.; Liu, X.; Fu, T.; et al. Population Structure and Selection Signal Analysis of Nanyang Cattle Based on Whole-Genome Sequencing Data. Genes 2024, 15, 351. [Google Scholar] [CrossRef] [PubMed]
  74. Luo, X.; Li, J.; Xiao, C.; Sun, L.; Xiang, W.; Chen, N.; Lei, C.; Lei, H.; Long, Y.; Long, T.; et al. Whole-Genome Resequencing of Xiangxi Cattle Identifies Genomic Diversity and Selection Signatures. Front. Genet. 2022, 13, 816379. [Google Scholar] [CrossRef] [PubMed]
  75. Shi, L.; Zhang, P.; Liu, Q.; Liu, C.; Cheng, L.; Yu, B.; Chen, H. Genome-Wide Analysis of Genetic Diversity and Selection Signatures in Zaobei Beef Cattle. Animals 2024, 14, 2447. [Google Scholar] [CrossRef]
  76. Xu, S.; Akhatayeva, Z.; Liu, J.; Feng, X.; Yu, Y.; Badaoui, B.; Esmailizadeh, A.; Kantanen, J.; Amills, M.; Lenstra, J.A.; et al. Genetic advancements and future directions in ruminant livestock breeding: From reference genomes to multiomics innovations. Sci. China Life Sci. 2024. [Google Scholar] [CrossRef]
  77. Wang, B.; Chen, L.; Wang, W. Genomic insights into ruminant evolution: From past to future prospects. Zool. Res. 2019, 40, 476–487. [Google Scholar] [CrossRef]
  78. Zhong, T.; Hou, D.; Zhao, Q.; Zhan, S.; Wang, L.; Li, L.; Zhang, H.; Zhao, W.; Yang, S.; Niu, L. Comparative Whole-Genome Resequencing to Uncover Selection Signatures Linked to Litter Size in Hu Sheep and Five Other Breeds. BMC Genom. 2024, 25, 480. [Google Scholar] [CrossRef]
  79. Zhao, L.; Yuan, L.; Li, F.; Zhang, X.; Tian, H.; Ma, Z.; Zhang, D.; Zhang, Y.; Zhao, Y.; Huang, K.; et al. Whole-Genome Resequencing of Hu Sheep Identifies Candidate Genes Associated with Agronomic Traits. J. Genet. Genom. 2024, 51, 866–876. [Google Scholar] [CrossRef]
  80. Yang, P.; Shang, M.; Bao, J.; Liu, T.; Xiong, J.; Huang, J.; Sun, J.; Zhang, L. Whole-Genome Resequencing Revealed Selective Signatures for Growth Traits in Hu and Gangba Sheep. Genes 2024, 15, 551. [Google Scholar] [CrossRef]
  81. YuYuan, C.; Lu, Z.; Guo, T.; Yue, Y.; Wang, X.; Wang, T.; Zhang, Y.; Hou, F.; Niu, C.; Sun, X.; et al. A Global Analysis of CNVs in Chinese Indigenous Fine-Wool Sheep Populations Using Whole-Genome Resequencing. BMC Genom. 2021, 22, 78. [Google Scholar] [CrossRef] [PubMed]
  82. Zhou, H.; Bai, L.; Li, S.; Li, W.; Wang, J.; Tao, J.; Hickford, J.G.H. Genetics of Wool and Cashmere Fibre: Progress, Challenges, and Future Research. Animals 2024, 14, 3228. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, H.; Zhu, S.; Guo, T.; Han, M.; Chen, B.; Qiao, G.; Wu, Y.; Yuan, C.; Liu, J.; Lu, Z.; et al. Whole-Genome Re-Sequencing Association Study on Yearling Wool Traits in Chinese Fine-Wool Sheep. J. Anim. Sci. 2021, 99, skab210. [Google Scholar] [CrossRef] [PubMed]
  84. Li, X.; Yang, J.; Shen, M.; Xie, X.-L.; Liu, G.-J.; Xu, Y.-X.; Lv, F.-H.; Yang, H.; Yang, Y.-L.; Liu, C.-B.; et al. Whole-Genome Resequencing of Wild and Domestic Sheep Identifies Genes Associated with Morphological and Agronomic Traits. Nat. Commun. 2020, 11, 2815. [Google Scholar] [CrossRef]
  85. Zhou, C.; Zhang, Y.; Ma, T.; Wu, D.; Yang, Y.; Wang, D.; Li, X.; Guo, S.; Yang, S.; Song, Y.; et al. Whole-Genome Resequencing of Ujimqin Sheep Identifies Genes Associated with Vertebral Number. Animals 2024, 14, 677. [Google Scholar] [CrossRef]
  86. Xiong, J.; Bao, J.; Hu, W.; Shang, M.; Zhang, L. Whole-Genome Resequencing Reveals Genetic Diversity and Selection Characteristics of Dairy Goat. Front. Genet. 2022, 13, 1044017. [Google Scholar] [CrossRef]
  87. Zhao, J.; Mu, Y.; Gong, P.; Liu, B.; Zhang, F.; Zhu, L.; Shi, C.; Lv, X.; Luo, J. Whole-Genome Resequencing of Native and Imported Dairy Goat Identifies Genes Associated with Productivity and Immunity. Front. Vet. Sci. 2024, 11, 1409282. [Google Scholar] [CrossRef]
  88. Fang, X.; Gu, B.; Chen, M.; Sun, R.; Zhang, J.; Zhao, L.; Zhao, Y. Genome-Wide Association Study of the Reproductive Traits of the Dazu Black Goat (Capra hircus) Using Whole-Genome Resequencing. Genes 2023, 14, 1960. [Google Scholar] [CrossRef]
  89. Li, J.; Xu, H.; Liu, X.; Xu, H.; Cai, Y.; Lan, X. Insight into the Possible Formation Mechanism of the Intersex Phenotype of Lanzhou Fat-Tailed Sheep Using Whole-Genome Resequencing. Animals 2020, 10, 944. [Google Scholar] [CrossRef]
  90. Wang, K.; Liu, X.; Qi, T.; Hui, Y.; Yan, H.; Qu, L.; Lan, X.; Pan, C. Whole-Genome Sequencing to Identify Candidate Genes for Litter Size and to Uncover the Variant Function in Goats (Capra hircus). Genomics 2021, 113 Pt 1, 142–150. [Google Scholar] [CrossRef]
  91. Guan, X.; Zhao, S.; Xiang, W.; Jin, H.; Chen, N.; Lei, C.; Jia, Y.; Xu, L. Genetic Diversity and Selective Signature in Dabieshan Cattle Revealed by Whole-Genome Resequencing. Biology 2022, 11, 1327. [Google Scholar] [CrossRef] [PubMed]
  92. Song, X.; Yao, Z.; Zhang, Z.; Lyu, S.; Chen, N.; Qi, X.; Liu, X.; Ma, W.; Wang, W.; Lei, C.; et al. Whole-Genome Sequencing Reveals Genomic Diversity and Selection Signatures in Xia’nan Cattle. BMC Genom. 2024, 25, 559. [Google Scholar] [CrossRef]
  93. Li, S.; Lei, H.; Li, J.; Sun, A.; Ahmed, Z.; Duan, H.; Chen, L.; Zhang, B.; Lei, C.; Yi, K. Analysis of Genetic Diversity and Selection Signals in Chaling Cattle of Southern China Using Whole-Genome Scan. Anim. Genet. 2023, 54, 284–294. [Google Scholar] [CrossRef] [PubMed]
  94. Dang, D.; Zhang, L.; Gao, L.; Peng, L.; Chen, J.; Yang, L. Analysis of Genomic Copy Number Variations through Whole-Genome Scan in Yunling Cattle. Front. Vet. Sci. 2024, 11, 1413504. [Google Scholar] [CrossRef] [PubMed]
  95. Shin, Y.; Jung, H.-J.; Jung, M.; Yoo, S.-I.; Subramaniyam, S.; Markkandan, K.; Kang, J.-M.; Rai, R.; Park, J.; Kim, J.-J. Discovery of Gene Sources for Economic Traits in Hanwoo by Whole-genome Resequencing. Asian-Australas. J. Anim. Sci. 2016, 29, 1353–1362. [Google Scholar] [CrossRef]
  96. Ahmad, S.F.; Chandrababu Shailaja, C.; Vaishnav, S.; Kumar, A.; Gaur, G.K.; Janga, S.C.; Ahmad, S.M.; Malla, W.A.; Dutt, T. Read-Depth Based Approach on Whole Genome Resequencing Data Reveals Important Insights into the Copy Number Variation (CNV) Map of Major Global Buffalo Breeds. BMC Genom. 2023, 24, 616. [Google Scholar] [CrossRef]
  97. Peng, W.; Fu, C.; Shu, S.; Wang, G.; Wang, H.; Yue, B.; Zhang, M.; Liu, X.; Liu, Y.; Zhang, J.; et al. Whole-Genome Resequencing of Major Populations Revealed Domestication-Related Genes in Yaks. BMC Genom. 2024, 25, 69. [Google Scholar] [CrossRef]
  98. Meng, G.; Bao, Q.; Ma, X.; Chu, M.; Huang, C.; Guo, X.; Liang, C.; Yan, P. Analysis of Copy Number Variation in the Whole Genome of Normal-Haired and Long-Haired Tianzhu White Yaks. Genes 2022, 13, 2405. [Google Scholar] [CrossRef]
  99. Wang, Y.; Cai, H.; Luo, X.; Ai, Y.; Jiang, M.; Wen, Y. Insight into Unique Somitogenesis of Yak (Bos grunniens) with One Additional Thoracic Vertebra. BMC Genom. 2020, 21, 201. [Google Scholar] [CrossRef]
  100. Henry, B.K.; Eckard, R.J.; Beauchemin, K.A. Review: Adaptation of ruminant livestock production systems to climate changes. Animal 2018, 12, s445–s456. [Google Scholar] [CrossRef]
  101. Jin, M.; Wang, H.; Liu, G.; Lu, J.; Yuan, Z.; Li, T.; Liu, E.; Lu, Z.; Du, L.; Wei, C. Whole-Genome Resequencing of Chinese Indigenous Sheep Provides Insight into the Genetic Basis Underlying Climate Adaptation. Genet. Sel. Evol. GSE 2024, 56, 26. [Google Scholar] [CrossRef] [PubMed]
  102. Hu, L.; Zhang, L.; Li, Q.; Liu, H.; Xu, T.; Zhao, N.; Han, X.; Xu, S.; Zhao, X.; Zhang, C. Genome-Wide Analysis of CNVs in Three Populations of Tibetan Sheep Using Whole-Genome Resequencing. Front. Genet. 2022, 13, 971464. [Google Scholar] [CrossRef] [PubMed]
  103. Zhou, C.; Cui, Y.; Yang, Y.; Guo, D.; Zhang, D.; Fan, Y.; Li, X.; Zou, J.; Xie, J. Runx1 Protects against the Pathological Progression of Osteoarthritis. Bone Res. 2021, 9, 50. [Google Scholar] [CrossRef] [PubMed]
  104. Huang, Z.; Chen, K.; Chi, Y.; Jin, H.; Li, L.; Zhang, W.; Xu, J.; Zhang, Y. Runx1 Regulates Zebrafish Neutrophil Maturation via Synergistic Interaction with C-Myb. J. Biol. Chem. 2021, 296, 100272. [Google Scholar] [CrossRef]
  105. Lee, S.H.; Manandhar, S.; Lee, Y.M. Roles of RUNX in Hypoxia-Induced Responses and Angiogenesis. Adv. Exp. Med. Biol. 2017, 962, 449–469. [Google Scholar]
  106. Wang, W.; Zhang, X.; Zhou, X.; Zhang, Y.; La, Y.; Zhang, Y.; Li, C.; Zhao, Y.; Li, F.; Liu, B.; et al. Deep Genome Resequencing Reveals Artificial and Natural Selection for Visual Deterioration, Plateau Adaptability and High Prolificacy in Chinese Domestic Sheep. Front. Genet. 2019, 10, 300. [Google Scholar] [CrossRef]
  107. Shen, J.; Hanif, Q.; Cao, Y.; Yu, Y.; Lei, C.; Zhang, G.; Zhao, Y. Whole Genome Scan and Selection Signatures for Climate Adaption in Yanbian Cattle. Front. Genet. 2020, 11, 94. [Google Scholar] [CrossRef]
  108. Buggiotti, L.; Yudin, N.S.; Larkin, D.M. Copy Number Variants in Two Northernmost Cattle Breeds Are Related to Their Adaptive Phenotypes. Genes 2022, 13, 1595. [Google Scholar] [CrossRef]
  109. Wang, T.; Ma, X.; Ma, C.; Wu, X.; Xi, T.; Yin, L.; Li, W.; Li, Y.; Liang, C.; Yan, P. Whole Genome Resequencing-Based Analysis of Plateau Adaptation in Meiren Yak (Bos grunniens). Anim. Biotechnol. 2024, 35, 2298406. [Google Scholar] [CrossRef]
  110. Lyu, Y.; Wang, F.; Cheng, H.; Han, J.; Dang, R.; Xia, X.; Wang, H.; Zhong, J.; Lenstra, J.A.; Zhang, H.; et al. Recent Selection and Introgression Facilitated High-Altitude Adaptation in Cattle. Sci. Bull. 2024, 69, 3415–3424. [Google Scholar] [CrossRef]
  111. Igoshin, A.; Yudin, N.; Aitnazarov, R.; Yurchenko, A.A.; Larkin, D.M. Whole-Genome Resequencing Points to Candidate DNA Loci Affecting Body Temperature under Cold Stress in Siberian Cattle Populations. Life 2021, 11, 959. [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. Zhong, Z.; Wang, Z.; Xie, X.; Pan, D.; Su, Z.; Fan, J.; Xiao, Q.; Sun, R. Insights into Adaption and Growth Evolution: Genome-Wide Copy Number Variation Analysis in Chinese Hainan Yellow Cattle Using Whole-Genome Re-Sequencing Data. Int. J. Mol. Sci. 2024, 25, 11919. [Google Scholar] [CrossRef] [PubMed]
  114. Xu, L.; Zhou, K.; Huang, X.; Chen, H.; Dong, H.; Chen, Q. Whole-Genome Resequencing Provides Insights into the Diversity and Adaptation to Desert Environment in Xinjiang Mongolian Cattle. BMC Genom. 2024, 25, 176. [Google Scholar] [CrossRef]
  115. Huang, N.; Zhao, L.; Wang, J.; Jiang, Q.; Ju, Z.; Wang, X.; Yang, C.; Gao, Y.; Wei, X.; Zhang, Y.; et al. Signatures of Selection in Indigenous Chinese Cattle Genomes Reveal Adaptive Genes and Genetic Variations to Cold Climate. J. Anim. Sci. 2023, 101, skad006. [Google Scholar] [CrossRef]
  116. Rafiepour, M.; Ebrahimie, E.; Vahidi, M.F.; Salekdeh, G.H.; Niazi, A.; Dadpasand, M.; Liang, D.; Si, J.; Ding, X.; Han, J.; et al. Whole-Genome Resequencing Reveals Adaptation Prior to the Divergence of Buffalo Subspecies. Genome Biol. Evol. 2021, 13, evaa231. [Google Scholar] [CrossRef]
  117. Sun, T.; Shen, J.; Achilli, A.; Chen, N.; Chen, Q.; Dang, R.; Zheng, Z.; Zhang, H.; Zhang, X.; Wang, S.; et al. Genomic Analyses Reveal Distinct Genetic Architectures and Selective Pressures in Buffaloes. GigaScience 2020, 9, giz166. [Google Scholar] [CrossRef]
  118. Surati, U.; Niranjan, S.K.; Pundir, R.K.; Koul, Y.; Vohra, V.; Gandham, R.K.; Kumar, A. Genome-Wide Comparative Analyses Highlight Selection Signatures Underlying Saline Adaptation in Chilika Buffalo. Physiol. Genom. 2024, 56, 609–620. [Google Scholar] [CrossRef]
  119. Bian, C.; Luo, Y.; Li, J.; Cheng, H.; He, F.; Duan, H.; Ahmed, Z.; Lei, C.; Yi, K. Inference of Genetic Diversity, Population Structure, and Selection Signatures in Xiangxi White Buffalo of China Through Whole-Genome Resequencing. Genes 2024, 15, 1450. [Google Scholar] [CrossRef]
  120. Place, S.E. Examining the role of ruminants in sustainable food systems. Grass Forage Sci. 2024, 79, 135–143. [Google Scholar] [CrossRef]
  121. Lyu, Y.; Yao, T.; Chen, Z.; Huangfu, R.; Cheng, H.; Ma, W.; Qi, X.; Li, F.; Chen, N.; Lei, C. Genomic characterization of dryland adaptation in endangered Anxi cattle in China. Anim. Genet. 2024, 55, 352–361. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, Q.; Lan, T.; Li, H.; Sahu, S.K.; Shi, M.; Zhu, Y.; Han, L.; Yang, S.; Li, Q.; Zhang, L.; et al. Whole-genome resequencing of Chinese pangolins reveals a population structure and provides insights into their conservation. Commun. Biol. 2022, 5, 821. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, R.; Wang, X.; Qi, Y.; Li, Y.; Na, Q.; Yuan, H.; Rong, Y.; Ao, X.; Guo, F.; Zhang, L.; et al. Genetic Diversity Analysis of Inner Mongolia Cashmere Goats (Erlangshan subtype) Based on Whole Genome Re-Sequencing. BMC Genom. 2024, 25, 698. [Google Scholar] [CrossRef]
  124. Sun, L.; Yuan, C.; Guo, T.; Zhang, M.; Bai, Y.; Lu, Z.; Liu, J. Resequencing Reveals Population Structure and Genetic Diversity in Tibetan Sheep. BMC Genom. 2024, 25, 906. [Google Scholar] [CrossRef] [PubMed]
  125. Luo, J.; Min, Q.; Sun, X.; Guo, X.; Song, M.; Zeng, X.; Guo, J.; Zhang, H.; Han, Y.; Li, L. Comparative Whole-Genome Analysis of Production Traits and Genetic Structure in Baiyu and Chuanzhong Black Goats. Animals 2024, 14, 3616. [Google Scholar] [CrossRef]
  126. Xu, L.; Zhang, Y.; Guo, Y.; Chen, Q.; Zhang, M.; Chen, H.; Geng, J.; Huang, X. Whole-Genome Resequencing Uncovers Diversity and Selective Sweep in Kazakh Cattle. Anim. Genet. 2024, 55, 377–386. [Google Scholar] [CrossRef]
  127. Sun, W.; Ren, H.; Li, M.; Mei, L.; Zhang, B.; Jia, X.; Chen, S.; Wang, J.; Lai, S. Genomic Insights and Conservation Priorities for Kongshan Cattle: A Whole-Genome Resequencing Approach. Animals 2024, 14, 3056. [Google Scholar] [CrossRef]
  128. Jin, L.; Qu, K.; Hanif, Q.; Zhang, J.; Liu, J.; Chen, N.; Suolang, Q.; Lei, C.; Huang, B. Whole-Genome Sequencing of Endangered Dengchuan Cattle Reveals Its Genomic Diversity and Selection Signatures. Front. Genet. 2022, 13, 833475. [Google Scholar] [CrossRef]
  129. Li, S.; Yu, J.; Kang, H.; Liu, J. Genomic Selection in Chinese Holsteins Using Regularized Regression Models for Feature Selection of Whole Genome Sequencing Data. Animals 2022, 12, 2419. [Google Scholar] [CrossRef]
  130. Zhou, D.K.; Zheng, Z.Q.; Yang, B.G.; Li, X.L.; Li, L.H.; Zhou, R.Y.; Nai, W.H.; Jiang, X.P.; Zhang, J.H.; Hong, Q.H.; et al. Identification of a Goat Intersexuality-Associated Novel Variant Through Genome-Wide Resequencing and Hi-C. Front. Genet. 2021, 11, 616743. [Google Scholar]
  131. Caballero, A.; Tenesa, A.; Keightley, P.D. The Nature of Genetic Variation for Complex Traits Revealed by GWAS and Regional Heritability Mapping Analyses. Genetics 2015, 201, 1601–1613. [Google Scholar] [CrossRef] [PubMed]
  132. Bekele, R.; Taye, M.; Abebe, G.; Meseret, S. Genomic Regions and Candidate Genes Associated with Milk Production Traits in Holstein and Its Crossbred Cattle: A Review. Int. J. Genom. 2023, 2023, 8497453. [Google Scholar] [CrossRef] [PubMed]
  133. Cheng, J.; Zhang, X.; Zhang, D.; Zhang, Y.; Li, X.; Zhao, Y.; Xu, D.; Zhao, L.; Li, W.; Wang, J.; et al. Sheep fecal transplantation affects growth performance in mouse models by altering gut microbiota. J. Anim. Sci. 2022, 100, skac303. [Google Scholar] [CrossRef] [PubMed]
  134. Zhao, B.; Luo, H.; Huang, X.; Wei, C.; Di, J.; Tian, Y.; Fu, X.; Li, B.; Liu, G.E.; Fang, L.; et al. Integration of a single-step genome-wide association study with a multi-tissue transcriptome analysis provides novel insights into the genetic basis of wool and weight traits in sheep. Genet. Sel. Evol. 2021, 53, 56. [Google Scholar] [CrossRef]
  135. Qi, G.; Si, Z.; Xuan, L.; Han, Z.; Hu, Y.; Fang, L.; Dai, F.; Zhang, T. Unravelling the genetic basis and regulation networks related to fibre quality improvement using chromosome segment substitution lines in cotton. Plant Biotechnol. J. 2024, 22, 3135–3150. [Google Scholar] [CrossRef]
  136. Jia, Y.; Shen, T.; Wen, Z.; Chen, J.; Liu, Q. Combining Transcriptome and Whole Genome Re-Sequencing to Screen Disease Resistance Genes for Wheat Dwarf Bunt. Int. J. Mol. Sci. 2023, 24, 17356. [Google Scholar] [CrossRef]
  137. Hu, Q.; Lian, Z.; Xia, X.; Tian, H.; Li, Z. Integrated chromatin accessibility and DNA methylation analysis to reveal the critical epigenetic modification and regulatory mechanism in gonadal differentiation of the sequentially hermaphroditic fish, Monopterus albus. Biol. Sex. Differ. 2022, 13, 73. [Google Scholar] [CrossRef]
  138. Horgusluoglu, E.; Neff, R.; Song, W.M.; Wang, M.; Wang, Q.; Arnold, M.; Krumsiek, J.; Galindo-Prieto, B.; Ming, C.; Nho, K.; et al. Integrative metabolomics-genomics approach reveals key metabolic pathways and regulators of Alzheimer’s disease. Alzheimers Dement. 2022, 18, 1260–1278. [Google Scholar] [CrossRef]
  139. Kumar, R.; Ruhel, R.; van Wijnen, A.J. Unlocking biological complexity: The role of machine learning in integrative multi-omics. Acad. Biol. 2024, 2, 1–3. [Google Scholar] [CrossRef]
  140. Bravo-Egana, V.; Sanders, H.; Chitnis, N. New Challenges, New Opportunities: Next Generation Sequencing and Its Place in the Advancement of HLA Typing. Hum. Immunol. 2021, 82, 478–487. [Google Scholar] [CrossRef]
  141. Deo, R.C. Machine Learning in Medicine. Circulation 2015, 132, 1920–1930. [Google Scholar] [CrossRef] [PubMed]
  142. Oyewole, G.J.; Thopil, G.A. Data Clustering: Application and Trends. Artif. Intell. Rev. 2023, 56, 6439–6475. [Google Scholar] [CrossRef]
  143. Alharbi, W.S.; Rashid, M. A Review of Deep Learning Applications in Human Genomics Using Next-Generation Sequencing Data. Hum. Genom. 2022, 16, 26. [Google Scholar] [CrossRef] [PubMed]
  144. Chen, C.; Wang, J.; Pan, D.; Wang, X.; Xu, Y.; Yan, J.; Wang, L.; Yang, X.; Yang, M.; Liu, G.-P. Applications of Multi-Omics Analysis in Human Diseases. MedComm 2023, 4, e315. [Google Scholar] [CrossRef]
  145. Aggarwal, V.; Banday, A.Z.; Jindal, A.K.; Das, J.; Rawat, A. Recent Advances in Elucidating the Genetics of Common Variable Immunodeficiency. Genes Dis. 2020, 7, 26–37. [Google Scholar] [CrossRef]
Table 1. Correspondence table of economic trait candidate genes.
Table 1. Correspondence table of economic trait candidate genes.
TraitsBovidaeCandidate GeneReference
BovinaCaprinae
ReproductionDabieshan Cattle, Huanhu yaks, Maiwa yaks, Yushu yaksHu Sheep, Saanen, Nubian, Alpine, Toggenburg, Guanzhong dairy goatBMPR1B, BMP2, PGFS, CYP19, CAMK4, GGT5, GNAQ, TSHR, TSHB, PTGS2, ESR2, ATP5E, AURKA, ENDOG, SOX2, RORA, GJA10, RXFP2, CDC25C, NANOS3, GPX5, GPX6, BTBD11, ARFIP1[79,86,87,90,91,97]
Vision Hu SheepALDH1A2, SAG, PDE6B[79]
ImmunityDabieshan Cattle, Xia’nan cattle, Huanhu yaks, Maiwa yaks, Yushu yaksHu Sheep, Saanen, Nubian, Alpine, Toggenburg, Guanzhong dairy goat, Xinong Saanen Dairy Goat, Laoshan Dairy GoatGPR35, SH2B2, PIK3R3, HRAS, JAK1, POU2F2, LRRC66, CTSZ, NELFCD, IGLL1, BOLA-DQA2, BOLA-DQB, IL11RA, CNTFR, CCL27, SLAMF1, SLAMF7, NAA35, GOLM1, INPP5D, ADCYAP1R1[79,86,87,91,92,97]
GrowthXia’nan cattle, Huanhu yaks, Maiwa yaks, Yushu yaksGangba sheep, Hu Sheep, Saanen, Nubian, Alpine, Toggenburg, Guanzhong dairy goat, Xinong Saanen Dairy Goat, Laoshan Dairy GoatHDAC1, YH7B, LCK, ACVR1, GNAI2, RBBP8, ACSL3, FBXW11, PLAT, CRB1, CTSZ, GHR, NR6A1, WSCD1, TMEM68, MFN1, NCKAP5, GRHL2, GRID2, SMARCAL1, EPHB2[80,87,92,97]
Yearling Staple Length Alpine Merino sheep, Chinese Merino sheep, Qinghai fine-wool sheep, Aohan fine-wool sheepFAM46A, LOC105607652, SGCD, PLCE1, RAB3C, AIM1, FAT3, C6H4orf22, PLA2R1, ANKRD42, KANSL2, ZNF804A, DPYD, LOC101114228, LOC101122569, LOC105607993, PLA2R1, LOC105611292, PRTFDC1, LOC101122517, C21H11orf85, BATF2[83]
Yearling Mean Fiber Diameter Alpine Merino sheep, Chinese Merino sheep, Qinghai fine-wool sheep, Aohan fine-wool sheepLOC106990409, ARID1A, MGMT, U2AF1, CRYAA, SLIT3, ARHGAP15, N4BP2, TRNAG-UCC, LOC105611432, LOC105603404, SLC4A11, LOC105603112, LOC105606895, LOC101115632[83]
Milk ProductionAnkole, Kenana, Huanhu yaks, Maiwa yaks, Yushu yaksSaanen, Nubian, Alpine, Toggenburg, Guanzhong dairy goat, Laoshan Dairy GoatGHR, DGAT2, ELF5, GLYCAM1, ACSBG2, ACSS2, STK3, PRELID3B, HECW1, HECW2, OSBPL2[86,87,97]
Udder Traits Dazu Black GoatATP1A1, LRRC4C, SPCS2, XRRA1, CELF4, NTM, TMEM45B, ATE1, FGFR2[88]
Litter Size Dazu Black Goat, Shaanbei White Cashmere GoatENSCHIG00000017110, SLC9A8, GLRB, GRIA2, GASK1B, ENSCHIG00000026285, CDC25C, ENDOG, NANOS3[88,90]
MeatXia’nan cattle, Huanhu yaks, Maiwa yaks, Yushu yaks MCCC1, BZW1, AOX1, LOC100138449, SPATA5, GRHL2[92,97]
Hair Growth And Hair-Follicle DevelopmentTianzhu white yaks ASTN2, ATM, COL22A1, GK5, SLIT3, PM20D1, SGCZ[98]
Table 2. Correspondence table of adaptive trait candidate genes.
Table 2. Correspondence table of adaptive trait candidate genes.
Adaptive TraitsBovidaeCandidate GeneReference
BovinaCaprinae
Drought-prone Kazakh sheep, Mongolian sheep, Tibetan sheep, Yunnan sheep, TBXT, TG, HOXA1[101]
High-altitude Kazakh sheep, Mongolian sheep, Tibetan sheep, Yunnan sheepDYSF, EPAS1, JAZF1, PDGFD, NF1[101]
HypoxiaTibetan cattleTibetan sheepRUNX1, CYP17, MBP2, ACVR2B, EGLN1[102,106,110]
Heat toleranceSiberian cattle, Lincang humped cattleDuolang sheep, Hainan black goat, Kazakh sheep, Mongolian sheep, Tibetan sheep, Yunnan sheep, DNAJB5, GNG2, MAPK8, CAPN2, SLC1A1, LEPR, GRIA4, COX17, MAATS1, UPK1B, IFNGR1, DDX23, PPT1, THBS1, CCL5, ATF1, PLA1A, PRKAG1, NR1I2, MED16, DNAJC8, HSPA4, FILIP1L, HELB, BCL2L1, TPX2, COMMD1, TSHR, ABCD4, TEX11[68,101,106,111]
Stress resistance Hainan black goatTLR2, IFI44, ENPP1, STK3, NFATC1[68]
Cold toleranceYanbian cattle, Tibetan cattle CORT, FGF5, CD36, LRP11, UQCR11, STING1, EGR1[107,110,115]
Water reabsorption and osmoregulation (desert adaptation)Xinjiang Mongolian cattle PDE11A, DIS3L2, SLIT2, KCNIP4, OSGEP, PKHD1, LOC617141, LOC112442378, LOC527385, FECH, LOC530929, LOC511936, CLIC4, FGF10[114]
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

Wang, Q.; Lu, Y.; Li, M.; Gao, Z.; Li, D.; Gao, Y.; Deng, W.; Wu, J. Leveraging Whole-Genome Resequencing to Uncover Genetic Diversity and Promote Conservation Strategies for Ruminants in Asia. Animals 2025, 15, 831. https://doi.org/10.3390/ani15060831

AMA Style

Wang Q, Lu Y, Li M, Gao Z, Li D, Gao Y, Deng W, Wu J. Leveraging Whole-Genome Resequencing to Uncover Genetic Diversity and Promote Conservation Strategies for Ruminants in Asia. Animals. 2025; 15(6):831. https://doi.org/10.3390/ani15060831

Chicago/Turabian Style

Wang, Qinqian, Ying Lu, Mengfei Li, Zhendong Gao, Dongfang Li, Yuyang Gao, Weidong Deng, and Jiao Wu. 2025. "Leveraging Whole-Genome Resequencing to Uncover Genetic Diversity and Promote Conservation Strategies for Ruminants in Asia" Animals 15, no. 6: 831. https://doi.org/10.3390/ani15060831

APA Style

Wang, Q., Lu, Y., Li, M., Gao, Z., Li, D., Gao, Y., Deng, W., & Wu, J. (2025). Leveraging Whole-Genome Resequencing to Uncover Genetic Diversity and Promote Conservation Strategies for Ruminants in Asia. Animals, 15(6), 831. https://doi.org/10.3390/ani15060831

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

Article Metrics

Back to TopTop