Go to Top Go to Bottom
Anim Biosci > Volume 38(12); 2025 > Article
Rong, Ao, Han, Xia, Shang, Lv, Wang, Su, Zhao, Zhang, and Wang: Whole-genome selection signatures identified candidate genes associated with cashmere traits in Inner Mongolia cashmere goats

Abstract

Objective

Inner Mongolia cashmere goats are superior indigenous breeds developed through long-term natural selection and systematic artificial selection, which have experienced a certain intensity of selection pressure during the breeding process, leading to bipolar differentiation trends in cashmere traits. Therefore, identifying genomic selection signatures associated with cashmere traits in Inner Mongolia cashmere goats is crucial for breeding high-quality cashmere-producing goats.

Methods

To unravel the genetic basis of cashmere traits, this study stratified 375 Inner Mongolia cashmere goats into eight subgroups based on breeding values for cashmere traits: high-yield vs low-yield cashmere types (HYCG vs LYCG), fine vs coarse cashmere types (FCG vs CCG), long vs short cashmere types (LCG vs SCG), and long vs short fleece types (LFCG vs SFCG). Whole-genome resequencing was performed for genotyping, followed by detection of selection signatures.

Results

Results revealed 144, 158, 147, and 147 high-frequency run of homozygosity (ROH) regions in HYCG, FCG, LCG, and LFCG subgroups, respectively, annotating to 515, 565, 510, and 521 genes. Additionally, genomic regions under positive selection were identified using FST, θπ ratios, and XP-EHH methods, with overlapping regions detected by ≥2 methods defined as candidate regions. Gene annotation identified 777, 660, 712, and 726 candidate genes in HYCG vs LYCG, FCG vs CCG, LCG vs SCG, and LFCG vs SFCG comparisons, respectively. These genes were enriched in 3,051 GO terms and 318 KEGG pathways, including Hippo, MAPK, Wnt, PI3K-Akt, and mTOR signaling pathways associated with cashmere growth and development, involving genes such as LGR6, RUNX2, IGF1R, FGF9, and TCF7L1.

Conclusion

In this study, we employed four complementary approaches, including ROHs, FST, θπ ratios, and XP-EHH, to identify genomic signatures of selection for cashmere traits in Inner Mongolia cashmere goats. These findings provide valuable insights for improving cashmere production performance and developing novel strains with high-quality cashmere in Inner Mongolia cashmere goats.

INTRODUCTION

As a major cashmere-producing region, the Inner Mongolia Autonomous Region is renowned for its Inner Mongolia cashmere goats, which are characterized by high cashmere yield, fine fiber diameter, and soft texture. Cashmere traits directly influence production efficiency and economic benefits in the cashmere goat industry. These traits represent complex polygenic characteristics controlled by multiple minor-effect loci, influenced by both genetic and environmental factors. Through prolonged natural and artificial selection, significant phenotypic divergence has emerged in cashmere traits among Inner Mongolia cashmere goat populations. During this selection process, nucleotide diversity and allele frequencies decreased in the genome of Inner Mongolia cashmere goats, while population differentiation and linkage disequilibrium increased and became stabilized within populations. These unique genetic footprints inscribed in the genome are referred to as selection signatures. Therefore, genome-wide scanning for these selection signatures and mapping candidate genes associated with cashmere traits will facilitate the development of genetic markers for genome-wide selective breeding programs in Inner Mongolia cashmere goats.
With the widespread application of whole-genome resequencing technology in livestock breeding, we now have sufficient information to identify selection signatures across the genome. Based on selection effects, methods for detecting selection signatures can be broadly categorized into four types: (1) Run of homozygosity (ROH) based on genomic heterozygosity; (2) Genetic differentiation index (FST) based on increased population divergence; (3) θπ ratios based on reduced nucleotide polymorphism; (4) Cross-Population Extended Haplotype Homozygosity (XP-EHH) based on increased linkage disequilibrium. Integrating all genomic feature information enables more accurate characterization of selection signatures across the genome associated with cashmere trait evolution. ROH refers to contiguous homozygous segments in the genome, whose formation is primarily associated with selection pressure and genetic drift [1]. Recently, ROH has been widely used as a powerful tool for identifying candidate genes and selection signatures related to economic traits in livestock [25]. ROH analysis can reveal genetic drift characteristics driven by selection pressure in Inner Mongolia cashmere goat populations and screen candidate genes associated with important economic traits. FST is a metric for measuring population differentiation caused by genetic structure [6], typically estimated from genetic polymorphism data such as single nucleotide polymorphisms (SNPs) or microsatellites. FST represents one of the most commonly used statistics in population genetics. θπ ratios evaluate genetic polymorphism indices by quantifying the proportion of nucleotide polymorphic sites. The XP-EHH method detects ongoing or nearly fixed selection signatures by comparing haplotypes between two populations [7].
To date, various genomic regions and candidate genes associated with cashmere traits have been identified using different approaches. Comparative genomics analysis revealed the critical roles of type IV collagen family genes (COL4A2, COL4A4, COL4A5, COL6A5, COL6A6) and integrin family genes (ITGA2, ITGA4, ITGA9, ITGB8) in cashmere trait formation [8]. FST and XP-EHH analyses identified key genes such as WNT10A, CSN3, TRPS1, and IGF1R in Inner Mongolia vs Liaoning cashmere goats, which may participate in hair follicle morphogenesis and stem cell pluripotency regulation [9]. Integrated multi-omics analysis of metabolome, transcriptome, and proteome demonstrated that DNMT3B, HMCN1, CPB2, GNG12, and other genes enriched in mTOR and amino acid metabolism pathways synergistically regulate cashmere fineness phenotypes [10]. RT-qPCR and immunofluorescence detection in skin tissues of Inner Mongolia cashmere goats (Arbas type) showed that FGF2, FGF21, and BMP7 exhibited peak expression at 3 months of age, potentially playing important regulatory roles in secondary hair follicle development [11]. Collectively, these studies indicate that cashmere traits are influenced by genetic factors. Despite these valuable findings, the genetic mechanisms underlying cashmere growth and development remain poorly understood. Therefore, identifying genes potentially involved in cashmere growth regulation continues to hold significant importance.
Since breeding values accurately reflect an individual’s genetic superiority, individuals with high breeding values are selected for reproduction during artificial selection, thereby improving the production performance of breeding populations. Therefore, this study stratified Inner Mongolia cashmere goats into contrasting groups based on breeding values: high-yield vs low-yield cashmere types, fine vs coarse cashmere types, long vs short cashmere types, and long vs short fleece types, to investigate genomic differences among these groups. Using SNP genotyping data, we identified high-frequency ROH regions by detecting runs of homozygosity in four subgroups: high-yield cashmere, fine cashmere, long cashmere, and long fleece types. Additionally, we measured FST, θπ ratios, and XP-EHH across eight subgroups to identify selection signatures in the genome of Inner Mongolia cashmere goats, thereby uncovering candidate genes associated with cashmere traits.

MATERIALS AND METHODS

Animal materials

Based on the calculated breeding values for four cashmere traits (cashmere yield, cashmere diameter, cashmere thickness, and fleece length) in Inner Mongolia cashmere goats [12], we defined eight subgroups: high-yield cashmere goats (HYCG), low-yield cashmere goats (LYCG), fine cashmere goats (FCG), coarse cashmere goats (CCG), long cashmere goats (LCG), short cashmere goats (SCG), long fleece cashmere goats (LFCG), and short fleece cashmere goats (SFCG). There were 100 individuals in each subgroup. The descriptive statistics of the phenotypic values of each subgroup are shown in Table 1. There are a total of 375 non-duplicate individuals in these 8 subgroups. Detailed sample information is provided in Supplement 1. Raw whole-genome resequencing data for the 375 Inner Mongolia cashmere goats have been deposited in the NCBI SRA database (PRJNA1096828).

Single nucleotide polymorphism calling

For data preprocessing, raw reads were first cleaned using fastp software (V0.20.0) [13]. Clean reads were then aligned to the goat reference genome (ARS1, GCF_001704415.1) using Burrows-Wheeler Aligner (BWA) software (V0.7.17) [14]. SAMtools software (V1.8–20) [15] was used to convert SAM files to BAM format and sort the BAM files. Genome Analysis Toolkit (GATK) (V3.8) [16] was employed for BAM file processing and SNP calling. After obtaining VCF files, VariantFiltration module was applied for filtering.

Genotype quality control and principal component analysis

To achieve high-quality SNPs, further quality control was performed using Plink (V1.90) [17] with the following criteria: (1) SNPs with call rate <98% were removed; (2) SNPs with minor allele frequency (MAF) <5% were excluded; (3) SNPs deviating from Hardy-Weinberg equilibrium (HWE) (p<10−6) were filtered out; (4) individuals with call rate <98% were excluded. The quality-controlled genotype data were visualized for SNP density distribution across chromosomes using the plot_snp_density() function in the R package HandyCNV (V1.1.6) [18]. The “--pca 3” parameter of Plink (V1.90) software was used to calculate the first three principal components of these eight subgroups, and the principal component analysis (PCA) plot was drawn using R (V3.6.0).

Runs of homozygosity detection

To identify genomic regions associated with ROHs in the four subgroups (high-yield cashmere, fine cashmere, long cashmere, and long fleece types), ROHs were detected using Plink software (V1.90). The following parameters were applied: sliding window size of 100 SNPs (--homozyg-window-snp 100); maximum 1 heterozygous SNP per window (--homozyg-window-het 1); maximum 1 missing SNP per window (--homozyg-window-missing 1); minimum SNP density of 1 per 50 kb (--homozyg-density 50); maximum gap between consecutive SNPs of 500 kb (--homozyg-gap 500); and minimum ROH length of 100 kb (--homozyg-kb 100).

Detection of high-frequency run of homozygosity regions

ROH results were summarized using the ‘roh_window ()’ function in the HandyCNV package [18]. A genomic sliding window size of 50 kb (window_size = 0.05) was applied for ROH visualization. ROH lengths were classified into five categories: 0.1–0.2 Mb, 0.2–0.4 Mb, 0.4–0.8 Mb, 0.8–1.6 Mb, and ≥1.6 Mb. Additionally, windows with sample frequency >25% in the four subgroups (high-yield cashmere, fine cashmere, long cashmere, and long fleece types) were defined as high-frequency ROH regions (threshold = 0.25). Adjacent windows exceeding the threshold were merged to generate a list of high-frequency ROH regions. The total autosomal length of the goat reference genome GCF_001704415.1 (ARS1) is approximately 2,466.191 Mb, which was used to calculate the proportion of ROH lengths across the genome.

Selection signature detection

To characterize the selected genomic regions in Inner Mongolia cashmere goats, we employed FST, θπ Ratio, and XP-EHH methods to detect genomic selection signatures across eight subgroups: HYCG vs LYCG, FCG vs CCG, LCG vs SCG, and LFCG vs SFCG. First, FST values between subgroup pairs (HYCG/LYCG, FCG/CCG, LCG/SCG, or LFCG/SFCG) were calculated using VCFtools (V0.1.16) [19] with a 50 kb sliding window and 10 kb step size. For θπ ratio calculations, VCFtools (V0.1.16) was used to compute θπ ratios between subgroups (LYCG/HYCG, CCG/FCG, SCG/LCG, or SFCG/LFCG) using the same 50 kb sliding window and 10 kb step size. For XP-EHH analysis, phasing of VCF files for 29 autosomes was performed using Beagle (V5.5), followed by XP-EHH value calculation with Selscan (V1.3.0) [20]. The top 1% of extreme values from each method were empirically selected as potential candidate regions under positive selection. Significant selection regions or SNPs were visualized with threshold lines in Manhattan plots using the CMplot package in R (V4.2.2) [21] (https://github.com/YinLiLin/R-CMplot, 20 January 2019).

Annotation and enrichment analysis of the candidate genes

Candidate regions identified by four complementary methods (ROHs, FST, θπ ratios, and XP-EHH) were annotated using Bedtools software (V2.30.0) [22]. For SNPs detected by XP-EHH, 50 kb upstream and downstream regions were annotated. The number of candidate genes from all methods was visualized using the Venny (V2.1) online tool (https://bioinfogp.cnb.csic.es/tools/venny/index.html). Additionally, Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed on candidate genes using the clusterProfiler package in R (V4.2.2) [23]. Manual curation of GO terms and KEGG signaling pathways was conducted to infer potential gene functions.

RESULTS

Single nucleotide polymorphism density statistical results

Following data quality control, each of the eight subgroups contained 100 individuals and 17,135,082 SNPs distributed across 29 autosomes, with an average density of 6,904 SNPs per megabase (Mb). Among these, 213 regions (1 Mb in length) exhibited SNP density >10,000 SNPs/Mb, 1 region had density <100 SNPs/Mb, and 2,268 regions showed density between 100–10,000 SNPs/Mb (Figure 1), indicating the high quality of our dataset.

Principal component analysis

We calculated the first three principal components by using Plink software, and the results showed that there is no group stratification among the eight subgroups (Figure 2). Therefore, the results of this study are not affected by the population genetic structure.

Run of homozygosity analysis

In the high-yield cashmere subgroup, a total of 59,385 ROHs were detected, with an average of 593.8 ROHs per sample and a maximum length of approximately 2.82 Mb. As the length of ROHs increased, the number of ROHs showed a downward trend. Among them, the number of ROHs in the 0.1–0.2 Mb length interval was the highest, accounting for about 72.8% of the total. On average, each sample had about 432.3 ROHs in this interval. The number of ROHs with a length of over 1.6 Mb was the lowest (Table 2). Additionally, 144 high - frequency ROHs (sample frequency ≥25%) were found, distributed across chromosomes 1–26 (Figure 3A). The total length of these high - frequency ROHs was about 25.9 Mb, accounting for approximately 1.05% of the total length of the autosomal genome. Among them, chromosomes 5 and 11 had the largest number of high - frequency ROH regions, with 14 high - frequency ROH regions detected on each. Moreover, there was a large region on chromosomes 2 and 18 where the frequency of ROH information was low, indicating a high level of genetic variability (Figure 3A). Among the 144 high - frequency ROHs, a total of 120 ROHs were annotated to 515 genes, while the remaining 24 ROHs had no annotations in the reference genome (Supplement 2).
In the fine cashmere subgroup, a total of 62,681 ROHs were detected, with the longest ROH measuring approximately 3.26 Mb. The number of ROHs decreased with increasing length, with the highest proportion (70.4% of total) observed in the 0.1–0.2 Mb interval (average 441.3 ROHs per sample), and the lowest proportion in ROHs ≥1.6 Mb (Table 3). Additionally, 158 high-frequency ROHs (sample frequency ≥25%) were identified (Figure 4A), spanning a total length of 28 Mb (1.13% of the autosomal genome). Chromosome 5 harbored the highest number of high-frequency ROH regions (16 regions). Notably, chromosomes 2 and 18 displayed large genomic regions with low ROH frequency, indicating a higher level of variation in this region (Figure 4A). Among the 158 high-frequency ROHs, 141 were annotated to 565 genes, while 17 ROHs had no annotations in the reference genome (Supplement 3).
In the long cashmere subgroup, a total of 59,071 ROHs were detected, with the maximum length of approximately 3.26 Mb. As the length of ROHs increased, the number of ROHs showed a downward trend. Among them, the number of ROHs in the 0.1–0.2 Mb length interval was the highest, accounting for about 73.3% of the total. On average, each sample had about 432.9 ROHs in this interval. The number of ROHs with a length of over 1.6 Mb was the lowest (Table 4). Additionally, 147 high - frequency ROHs (sample frequency ≥25%) were found, distributed across chromosomes 1–26 (Figure 5A). The total length of these high - frequency ROHs was about 23.8 Mb, accounting for approximately 0.97% of the total length of the autosomal genome. Among them, chromosomes 5 and 11 had the largest number of high - frequency ROH regions, with 14 high - frequency ROH regions detected on each. Moreover, there was a large region on chromosomes 2 and 18 where the frequency of ROH information was low, indicating a high level of genetic variability (Figure 5A). Among the 147 high - frequency ROHs, a total of 123 ROHs were annotated to 510 genes, while the remaining 24 ROHs had no annotations in the reference genome (Supplement 4).
In the long fleece subgroup, a total of 56,856 ROHs were detected, with the maximum length of approximately 2.57 Mb. As the length of ROHs increased, the number of ROHs showed a downward trend. Among them, the number of ROHs in the 0.1–0.2 Mb length interval was the highest, accounting for about 74.3% of the total. On average, each sample had about 422.6 ROHs in this interval. The number of ROHs with a length of over 1.6 Mb was the lowest (Table 5). Additionally, 149 high - frequency ROHs (sample frequency ≥25%) were found, distributed across chromosomes 1–26 (Figure 6A). The total length of these high - frequency ROHs was about 25.1 Mb, accounting for approximately 1.02% of the total length of the autosomal genome. Among them, chromosomes 5 and 11 had the largest number of high - frequency ROH regions, with 14 high - frequency ROH regions detected on each. Moreover, there were two large genomic regions on chromosome 2 where ROH frequency was low, indicating high genetic variability (Figure 6A). Among the 147 high - frequency ROHs, a total of 127 ROHs were annotated to 521 genes, while the remaining 20 ROHs had no annotations in the reference genome (Supplement 5).

Genome-wide scan of selection signature detection

HYCG VS LYCG

We calculated FST, θπ ratios, and XP-EHH values between HYCG and LYCG subgroups, using the top 1% thresholds (FST≥0.02, θπ ratios≥1.14, XP-EHH≥0.34) to identify significant selection regions. The FST method detected 2,466 selection regions in HYCG, annotated to 753 genes (Figure 3B, Supplement 6). The θπ ratios method also identified 2,466 selection regions, annotated to 878 genes (Figure 3C, Supplement 7). The XP-EHH method detected 171,305 candidate SNPs, annotated to 3,527 genes (Figure 3D, Supplement 8). Additionally, 144 high-frequency ROHs were identified in HYCG, annotating to 515 candidate genes. A total of 777 shared genes were detected by two or more methods, with 10 genes including MYCBP2, DPY19L4, and INTS8 identified by all four methods (Figure 3E).

FCG VS CCG

By calculating FST, θπ ratios, and XP-EHH values between FCG and CCG subgroups, we identified genomic selection signatures in the FCG subgroup during artificial selection. The top 1% thresholds (FST≥0.02, θπ ratios≥1.15, XP-EHH≥0.29) were used to identify selection regions. The FST method detected 2,466 selection regions in FCG, annotated to 748 genes (Figure 4B, Supplement 9). The θπ ratios method also identified 2,466 selection regions, annotated to 873 genes (Figure 4C, Supplement 10). The XP-EHH method detected 171,308 candidate SNPs, annotated to 3,461 genes (Figure 4D, Supplement 11). Additionally, 158 high-frequency ROHs were identified in FCG, annotating to 565 candidate genes. A total of 660 shared genes were detected by two or more methods, with LOC106501971 identified by all four methods (Figure 4E).

LCG VS SCG

By calculating FST, θπ ratios, and XP-EHH values between LCG and SCG subgroups, we identified genomic selection signatures in the LCG subgroup during artificial selection. The top 1% thresholds (FST≥0.02, θπ ratios≥1.14, XP-EHH ≥0.29) were used to identify selection regions. The FST method detected 2,466 selection regions in LCG, annotated to 600 genes (Figure 5B, Supplement 12). The θπ ratios method also identified 2,466 selection regions, annotated to 814 genes (Figure 5C, Supplement 13). The XP-EHH method detected 171,308 candidate SNPs, annotated to 4,014 genes (Figure 5D, Supplement 14). Additionally, 147 high-frequency ROHs were identified in LCG, annotating to 510 candidate genes. A total of 712 shared genes were detected by two or more methods, with nine genes including VRK2, FANCL, and FRY identified by all four methods (Figure 5E).

LFCG VS SFCG

By calculating FST, θπ ratios, and XP-EHH values between LFCG and SFCG subgroups, we identified genomic selection signatures in the LFCG subgroup during artificial selection. The top 1% thresholds (FST≥0.01, θπ ratios≥1.13, XP-EHH≥0.32) were used to identify selection regions. The FST method detected 2,466 selection regions in LFCG, annotated to 741 genes (Figure 6B, Supplement 15). The θπ ratios method also identified 2,466 selection regions, annotated to 859 genes (Figure 6C, Supplement 16). The XP-EHH method detected 171,308 candidate SNPs, annotated to 3,570 genes (Figure 6D, Supplement 17). Additionally, 147 high-frequency ROHs were identified in LFCG, annotating to 521 candidate genes. A total of 726 shared genes were detected by two or more methods, with 22 genes including MYCBP2, DPY19L4, and INTS8 identified by all four methods (Figure 6E).

Enrichment analysis of candidate gene

A total of 2,261 candidate genes were detected across eight Inner Mongolia cashmere goat subgroups using two or more methods (Figure 7). Based on these genes, we further screened out the top candidate genes with definite functions in hair follicle biology, as shown in Table 6. Functional annotation and enrichment analysis of these genes were performed using the clusterProfiler package, revealing enrichment in 3,051 GO terms and 318 KEGG pathways (Supplements 18, 19). Enriched GO terms included cell-cell junction (GO:0005911), antioxidant activity (GO:0016209), cell adhesion (GO:0007155), fibroblast growth factor receptor binding (GO:0005104), and cell cycle process (GO:0022402) (Figure 8A), which are associated with cashmere growth and development [2427]. KEGG pathways such as ABC transporters (chx02010), Hippo (chx04390), cAMP (chx04024), MAPK (chx04010), Ras (chx04014), and Wnt (chx04310) were also enriched (Figure 8B) [28]. Notably, 27 shared genes were identified across all eight subgroups. Among them, RELN was enriched in focal adhesion (chx04510), ECM-receptor interaction (chx04512), and PI3K-Akt signaling pathway (chx04151); TAOK1 was enriched in MAPK (chx04010); CCNE2 was involved in cell cycle process (GO:0022402) and PI3K-Akt (chx04151); and SEH1L and INTS8 were enriched in mTOR (chx04150) signaling pathway. These pathways are all associated with cashmere growth and development.

DISCUSSION

In this study, we first detected ROHs in four target groups: high-yield cashmere, fine cashmere, long cashmere, and long fleece types. The length of ROHs reflects a population’s inbreeding history, as the probability of ROH disruption by recombination increases with generations. Shorter ROHs typically result from ancient inbreeding, while longer ROHs indicate recent inbreeding events [29]. We found that ROH lengths in the four target subgroups (high-yield cashmere, fine cashmere, long cashmere, and long fleece types) were concentrated in the 0.1–0.2 Mb interval. These shorter ROHs suggest no severe inbreeding occurred in recent generations of Inner Mongolia cashmere goats. During selection, the rapid increase in frequency of haplotypes carrying beneficial mutations can also increase ROH number and length, leading to reduced genetic variation in flanking regions of target loci. Thus, ROHs are regarded as genomic footprints of recent positive selection [30]. Therefore, ROHs can be used to identify selection signals and map candidate genes. In this study, gene annotation of high-frequency ROH regions identified 515, 565, 510, and 521 candidate genes in the four target groups.
To identify candidate genes associated with cashmere traits more accurately, we employed FST, θπ ratios, and XP-EHH methods for genomic selection signature analysis in Inner Mongolia cashmere goats. A total of 777, 660, 712, and 726 candidate genes (detected by two or more methods) were identified in HYCG vs LYCG, FCG vs CCG, LCG vs SCG, and LFCG vs SFCG comparisons, respectively. Among them, 27 candidate genes were commonly detected across all populations, suggesting their potential regulatory roles in cashmere trait selection. Studies have shown that LGR6, as a biomarker of isthmic stem cells in hair follicles, is enriched in the Wnt signaling pathway and can induce stem cells to differentiate into hair follicles, sebaceous glands and epidermal cells, participating in multilineage differentiation and tissue repair of hair follicles [31,32]. RUNX2 is enriched in the Wnt and Hedgehog signaling pathways, expressed in the dermal papillae and epithelial cells of developing hair follicles, and affects hair follicle morphogenesis and cycle regulation [33]. FGF9 is enriched in the FGF signaling pathway, expressed in dermal papillary cells, and promotes the activation of hair follicle stem cells and hair follicle regeneration [34]. IGF1R is enriched in the IGF signaling pathway, expressed in dermal papillary cells, participates in the signal transduction of cell proliferation and survival, and plays an important role in maintaining the normal function of dermal papillary cells and hair follicle development [35,36]. TCF7L1 is enriched in the Wnt signaling pathway and, as a key transcription factor of the Wnt signaling pathway, participates in regulating processes such as cell proliferation and differentiation [37]. After activation of the PI3K-Akt and mTOR signaling pathways, it can promote the proliferation, migration and cell cycle progression of human hair follicle dermal papilla cells, and simultaneously activate the Wnt/β-catenin pathway, jointly promoting hair follicle growth [38]. Another contrary study showed that corticotropin-releasing hormone inhibited autophagy of hair follicle dermal papilla cells and promoted apoptosis by suppressing PTEN in the PI3K/AKT/mTOR signaling pathway, resulting in blocked hair follicle regeneration and triggering stress-induced alopecia [39]. Researchers can simultaneously activate the Wnt/β-catenin and MAPK signaling pathways through alternating current stimulation. The two jointly promote the proliferation of human dermal papillary cells and the expression of hair follicle-related genes (such as KGF and VEGF). The regulation of this synergistic effect on hair follicle growth and hair growth cycle was further verified in the rabbit model [40]. Collectively, these candidate genes play crucial roles in regulating cashmere growth and development, offering valuable insights for improving cashmere production performance in Inner Mongolia cashmere goats.

CONCLUSION

In this study, we employed four complementary approaches, including ROHs, FST, θπ ratios, and XP-EHH, to identify genomic signatures of selection for cashmere traits in Inner Mongolia cashmere goats. Numerous candidate regions and genes were identified, such as LGR6, RUNX2, IGF1R, FGF9, and TCF7L1, with most genes associated with hair follicle morphogenesis and cashmere growth and development. Our findings contribute to a better understanding of the breeding potential and genetic uniqueness of Inner Mongolia cashmere goats, and are of significant importance for elucidating the genetic mechanisms underlying cashmere trait formation.

Notes

CONFLICT OF INTEREST

No potential conflict of interest relevant to this article was reported.

AUTHORS’ CONTRIBUTION

Conceptualization: Zhang Y, Wang R.

Data curation: Rong Y, Ao X, Han M.

Formal analysis: Rong Y.

Methodology: Xia Q.

Writing - original draft: Rong Y.

Writing - review & editing: Rong Y, Ao X, Han M, Xia Q, Shang F, Lv Q, Wang Z, Su R, Zhao Y, Zhang Y, Wang R.

FUNDING

This work was supported by the Science and Program of Inner Mongolia Autonomous Region (2025KYPT0103); National Key Research and Development Program of China (2021YFD1200902); Major Science and Technology Program of Inner Mongolia Autonomous Region (2021ZD0012); Inner Mongolia Education Department Special Research Project for First Class Disciplines (YLXKZX-NND-007); Program for Innovative Research Team in Universities of the Inner Mongolia Autonomous Region (NMGIRT2322); Basic Scientific Research Business Fee Project for Universities Directly under the Inner Mongolia Autonomous Region (BR251201); Inner Mongolia Autonomous Region Joint Breeding Research Project-"Innovation in Goat and Sheep Genetic Resources and Breeding System Construction" (YZ2023011); The Project of Northern Agriculture and Livestock Husbandry Technical Innovation Center, Chinese Academy of Agricultural Sciences (BFGJ2022002).

ACKNOWLEDGMENTS

Thanks to all the staff of Erlangshan Ranch of Inner Mongolia Beiping Textile Co., Ltd. for their contributions in the production performance measurement.

DATA AVAILABILITY

The datasets generated and analyzed during the current study are available within the article. The data that support the findings of this study can be obtained from the College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China (whole genome re-sequence data). However, restrictions apply to the availability of these data, as they were used under a license for this study. Access to the data is available from the corresponding authors with the permission of Inner Mongolia Agricultural University.

ETHICS APPROVAL

All experiments and procedures were carried out following the Scientific Research and Academic Ethics Committee of Inner Mongolia Agricultural University and the Biomedical Research Ethics of Inner Mongolia Agricultural University (Approval No. [2020] 056).

DECLARATION OF GENERATIVE AI

No AI tools were used in this article.

SUPPLEMENTARY MATERIAL

Supplementary file is available from: https://doi.org/10.5713/ab.25.0252
Supplement 1. Sample information for 8 subpopulations in IMCGs.
ab-25-0252-Supplementary-1.xlsx
Supplement 2. High-frequency ROH regions and annotated genes in HYCG.
ab-25-0252-Supplementary-2.pdf
Supplement 3. High-frequency ROH regions and annotated genes in FCG.
ab-25-0252-Supplementary-3.pdf
Supplement 4. High-frequency ROH regions and annotated genes in LCG.
ab-25-0252-Supplementary-4.pdf
Supplement 5. High-frequency ROH regions and annotated genes in LFCG.
ab-25-0252-Supplementary-5.pdf
Supplement 6. Detected and gene annotation of candidate regions by FST (HYCG VS LYCG).
ab-25-0252-Supplementary-6.pdf
Supplement 7. Detected and gene annotation of candidate regions by θπ ratios (LYCG VS HYCG).
ab-25-0252-Supplementary-7.pdf
Supplement 8. Detected and gene annotation of candidate regions by XP-EHH (HYCG VS LYCG).
Supplement 9. Detected and gene annotation of candidate regions by FST (FCG VS CCG).
Supplement 10. Detected and gene annotation of candidate regions by θπ ratios (CCG VS FCG).
Supplement 11. Detected and gene annotation of candidate regions by XP-EHH (FCG VS CCG).
Supplement 12. Detected and gene annotation of candidate regions by FST (LCG VS SCG).
Supplement 13. Detected and gene annotation of candidate regions by θπ ratios (SCG VS LCG).
Supplement 14. Detected and gene annotation of candidate regions by XP-EHH (LCG VS SCG).
Supplement 15. Detected and gene annotation of candidate regions by FST (LFCG VS SFCG).
Supplement 16. Detected and gene annotation of candidate regions by θπ ratios (SFCG VS LFCG).
Supplement 17. Detected and gene annotation of candidate regions by XP-EHH (LFCG VS SFCG).
Supplement 18. GO enrichment analysis results of candidate genes in 8 Inner Mongolia cashmere goats subgroup.
Supplement 19. KEGG enrichment analysis results of candidate genes in 8 Inner Mongolia cashmere goats subgroup.
ab-25-0252-Supplementary-8-19.pdf

Figure 1
Whole-genome SNP density distribution in Inner Mongolia cashmere goats. Regions with SNP density ≤100 are represented by purple dots, regions with density between 100–10,000 are shown as pink dots, and regions with density ≥10,000 are indicated by green dots. SNP, single nucleotide polymorphism.
ab-25-0252f1.jpg
Figure 2
Visualization of PCA. The first three explain the variance percentage (PC1, PC2, and PC3) as the X, Y, and Z axes. (A) represents the PCA visualization of HYCG and LYCG. (B) represents the PCA visualization of CCG and FCG. (C) represents the PCA visualization of LCG and SCG. (D) represents the PCA visualization of LFCG and SFCG. HYCG, high-yield cashmere goats; LYCG, low-yield cashmere goats; CCG, coarse cashmere goats; FCG, fine cashmere goats; LCG, long cashmere goats; SCG, short cashmere goats; LFCG, long fleece cashmere goats; SFCG, short fleece cashmere goats; PCA, principal component analysis.
ab-25-0252f2.jpg
Figure 3
Genome-wide selection scan in high-yield cashmere goats (HYCG) and low-yield cashmere goats (LYCG). Using sliding window analysis (50 kb window size, 10 kb step size). (A) High-frequency ROH Regions. (B) Selection signatures for FST. (C) Selection signatures for θπ ratios. (D) Selection signatures for XP-EHH. (E) The number of genes overlapping with each other among the results of four detection methods, and the genes detected simultaneously by all four methods. Threshold (top 1%) of FST, θπ ratios, XP-EHH is marked with a horizontal red line. ROH, run of homozygosity.
ab-25-0252f3.jpg
Figure 4
Genome-wide selection scan in fine cashmere goats (FCG) and coarse cashmere goats (CCG). Using sliding window analysis (50 kb window size, 10 kb step size). (A) High-frequency ROH Regions. (B) Selection signatures for FST. (C) Selection signatures for θπ ratios. (D) Selection signatures for XP-EHH. (E) The number of genes overlapping with each other among the results of four detection methods, and the genes detected simultaneously by all four methods. Threshold (top 1%) of FST, θπ ratios, XP-EHH is marked with a horizontal red line. ROH, run of homozygosity.
ab-25-0252f4.jpg
Figure 5
Genome-wide selection scan in long cashmere goats (LCG) and short cashmere goats (SCG). Using sliding window analysis (50 kb window size, 10 kb step size). (A) High-frequency ROH Regions. (B) Selection signatures for FST. (C) Selection signatures for θπ ratios. (D) Selection signatures for XP-EHH. (E) The number of genes overlapping with each other among the results of four detection methods, and the genes detected simultaneously by all four methods. Threshold (top 1%) of FST, θπ ratios, XP-EHH is marked with a horizontal red line. ROH, run of homozygosity.
ab-25-0252f5.jpg
Figure 6
Genome-wide selection scan in long fleece cashmere goats (LFCG) and short fleece cashmere goats (SFCG). Using sliding window analysis (50 kb window size, 10 kb step size). (A) High-frequency ROH Regions. (B) Selection signatures for FST. (C) Selection signatures for θπ ratios. (D) Selection signatures for XP-EHH. (E) The number of genes overlapping with each other among the results of four detection methods, and the genes detected simultaneously by all four methods. Threshold (top 1%) of FST, θπ ratios, XP-EHH is marked with a horizontal red line. ROH, run of homozygosity.
ab-25-0252f6.jpg
Figure 7
Overlapping selection candidate genes by ROHs, FST, θπ ratios, and XP-EHH in HYCG/LYCG, FCG/CCG, LCG/SCG and LFCG/SFCG. FCG, fine cashmere goats; CCG, coarse cashmere goats; LCG, long cashmere goats; SCG, short cashmere goats; HYCG, high-yield cashmere goats; LYCG, low-yield cashmere goats; ROH, run of homozygosity; LFCG, long fleece cashmere goats; SFCG, short fleece cashmere goats.
ab-25-0252f7.jpg
Figure 8
Bubble diagram of GO and KEGG enrichment analysis of the candidate genes. (A) represents the GO enrichment results, in the legend, BP represents biological process, CC represents cellular component, MF represents molecular function. (B) represents the KEGG enrichment results.
ab-25-0252f8.jpg
Table 1
Descriptive statistics of phenotypic values of 8 subgroups
Subgroups Number of records Max Mean Min SD CV (%)
HYCG 100 544.9 190.1799 86.64 91.89 48.32
LYCG 100 −103.75 −179.837 −727.42 96.70 −53.77
CCG 100 1.99 0.8763 0.45 0.38 43.64
FCG 100 −0.31 −0.6599 −2.56 0.38 −58.29
LCG 100 2.23 0.8342 0.42 0.35 41.99
SCG 100 −0.37 −0.7179 −1.9 0.32 −44.69
LFCG 100 5.99 2.5948 1.34 1.05 40.63
SFCG 100 −1.07 −2.5264 −6.21 1.20 −47.41

HYCG, high-yield cashmere goats; LYCG, low-yield cashmere goats; CCG, coarse cashmere goats; FCG, fine cashmere goats; LCG, long cashmere goats; SCG, short cashmere goats; LFCG, long fleece cashmere goats; SFCG, short fleece cashmere goats.

Table 2
ROH statistics in different length regions in HYCG
Length_group (Mb) Number_ROH Mean SD Min Max Proportion N_per_animal
0.1–0.2 43,233 134,476 26,779 100,000 199,998 0.728 432.3
0.2–0.4 12,273 268,820 53,806 200,002 399,975 0.207 122.7
0.4–0.8 3,297 526,140 102,420 400,014 799,560 0.056 33
0.8–1.6 553 1,004,979 175,249 800,033 1,594,910 0.009 5.5
≥1.6 29 2,012,450 326,898 1,610,159 2,823,986 0 0.3
Total 59,385 593.8

ROH, run of homozygosity; HYCG, high-yield cashmere goats; SD, standard deviation.

Table 3
ROH statistics in different length regions in FCG
Length_group (Mb) Number_ROH Mean SD Min Max Proportion N_per_animal
0.1–0.2 44,130 135,062 26,988 100,001 199,998 0.704 441.3
0.2–0.4 13,672 271,104 54,849 200,004 399,999 0.218 136.7
0.4–0.8 4,170 531,446 104,552 400,010 799,793 0.067 41.7
0.8–1.6 672 1,001,052 167,797 800,025 1,572,122 0.011 6.7
≥1.6 37 1,912,380 361,752 1,600,830 3,258,204 0.001 0.4
Total 62,681 626.8

ROH, run of homozygosity; FCG, fine cashmere goats; SD, standard deviation.

Table 4
ROH statistics in different length regions in LCG
Length_group (Mb) Number_ROH Mean SD Min Max Proportion N_per_animal
0.1–0.2 43,288 134,438 26,751 100,000 199,953 0.733 432.9
0.2–0.4 12,128 268,578 53,936 200,009 399,656 0.205 121.3
0.4–0.8 3,160 525,122 101,031 400,010 799,973 0.053 31.6
0.8–1.6 472 1,001,740 172,745 800,760 1,597,842 0.008 4.7
≥1.6 23 1,887,542 350,081 1,602,934 3,184,378 0 0.2
Total 59,071 590.7

ROH, run of homozygosity; LCG, long cashmere goats; SD, standard deviation.

Table 5
ROH statistics in different length regions in LFCG
Length_group (Mb) Number_ROH Mean SD Min Max Proportion N_per_animal
0.1–0.2 42,256 134,338 26,816 100,000 199,993 0.743 422.6
0.2–0.4 11,261 267,044 53,410 200,009 399,987 0.198 112.6
0.4–0.8 2,867 525,337 100,026 400,010 797,377 0.05 28.7
0.8–1.6 448 999,405 164,558 800,892 1,597,842 0.008 4.5
≥1.6 24 1,943,122 282,111 1,612,312 2,571,696 0 0.2
Total 56,856 568.6

ROH, run of homozygosity; LFCG, long fleece cashmere goats; SD, standard deviation.

Table 6
The functions of candidate genes in hair follicle biology
Chromosomal region (Mb) Genes Methods Functional association References
16:78.85–78.90 LGR6 FST, XP-EHH It is expressed in the stem cells located in the central isthmus of hair follicles, and participates in the cell differentiation and regeneration of hair follicles and epidermis. [31,32]
23:30.04–30.10 RUNX2 ROH, FST, XP-EHH This gene exhibits dynamic stage-specific expression in hair follicles, and its deletion delays hair follicle maturation, leading to significant reduction in skin and epidermal thickness and weakened epidermal cell proliferation. [33]
12:50.25–51.40 FGF9 ROH, θπ ratios FGF9 can promote the transition of hair follicles from telogen to anagen, accelerate the proliferation of dermal papilla cells, and thus induce hair regeneration by regulating the Wnt/β-catenin signaling pathway. [34]
21:7.08–7.18 IGF1R FST, XP-EHH As a target gene of miR-1 and miR-let7a, its expression level is higher in the anagen phase than in the catagen phase of hair follicles in Liaoning cashmere goats, and it participates in the regulation of proliferation and differentiation of hair follicle stem cells through the IGF signaling pathway. [35,36]
11:49.38–49.48 TCF7L1 FST, XP-EHH As a key transcription factor of the Wnt signaling pathway, the down-regulation of this gene’s expression in canine skin leads to the obstruction of hair follicle stem cell activation, and then participates in regulating the transition process of hair follicles from the resting phase to the anagen phase. [37]

ROH, run of homozygosity.

REFERENCES

1. Gibson J, Morton NE, Collins A. Extended tracts of homozygosity in outbred human populations. Hum Mol Genet 2006;15:789–95. https://doi.org/10.1093/hmg/ddi493
crossref pmid
2. Yin C, Wang Y, Zhou P, et al. Genomic scan for runs of homozygosity and selective signature analysis to identify candidate genes in large white pigs. Int J Mol Sci 2023;24:12914. https://doi.org/10.3390/ijms241612914
crossref pmid pmc
3. Lukic B, Curik I, Drzaic I, et al. Genomic signatures of selection, local adaptation and production type characterisation of East Adriatic sheep breeds. J Anim Sci Biotechnol 2023;14:142. https://doi.org/10.1186/s40104-023-00936-y
crossref pmid pmc
4. Ramljak J, Špehar M, Ceranac D, et al. Genomic characterization of local croatian sheep breeds-effective population size, inbreeding & signatures of selection. Animals 2024;14:1928. https://doi.org/10.3390/ani14131928
crossref pmid pmc
5. Bao J, Xiong J, Huang J, Yang P, Shang M, Zhang L. Genetic diversity, selection signatures, and genome-wide association study identify candidate genes related to litter size in Hu sheep. Int J Mol Sci 2024;25:9397. https://doi.org/10.3390/ijms25179397
crossref pmid pmc
6. Weir BS, Cockerham CC. Estimating F-statistics for the analysis of population structure. Evolution 1984;38:1358–70. https://doi.org/10.1111/j.1558-5646.1984.tb05657.x
crossref pmid
7. Sabeti PC, Varilly P, Fry B, et al. Genome-wide detection and characterization of positive selection in human populations. Nature 2007;449:913–8. https://doi.org/10.1038/nature06250
crossref pmid pmc
8. Wang W, Li Z, Xie G, et al. Convergent genomic signatures of cashmere traits: evidence for natural and artificial selection. Int J Mol Sci 2023;24:1165. https://doi.org/10.3390/ijms24021165
crossref pmid pmc
9. Jin M, Lu J, Fei X, et al. Genetic signatures of selection for cashmere traits in Chinese goats. Animals 2020;10:1905. https://doi.org/10.3390/ani10101905
crossref pmid pmc
10. Diao X, Yao L, Wang X, et al. Hair follicle development and cashmere traits in Albas goat kids. Animals 2023;13:617. https://doi.org/10.3390/ani13040617
crossref pmid pmc
11. Zhao B, Suo L, Wu Y, et al. Stress adaptation in Tibetan cashmere goats is governed by inherent metabolic differences and manifested through variable cashmere phenotypes. Genomics 2024;116:110801. https://doi.org/10.1016/j.ygeno.2024.110801
crossref pmid
12. Rong Y, Wang X, Na Q, et al. Genome-wide association study for cashmere traits in Inner Mongolia cashmere goat population reveals new candidate genes and haplotypes. BMC Genomics 2024;25:658. https://doi.org/10.1186/s12864-024-10543-4
crossref pmid pmc
13. Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 2018;34:i884–90. https://doi.org/10.1093/bioinformatics/bty560
crossref pmid pmc
14. Li H, Durbin R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 2009;25:1754–60. https://doi.org/10.1093/bioinformatics/btp324
crossref pmid pmc
15. Li H, Handsaker B, Wysoker A, et al. The sequence alignment/map format and SAMtools. Bioinformatics 2009;25:2078–9. https://doi.org/10.1093/bioinformatics/btp352
crossref pmid pmc
16. McKenna A, Hanna M, Banks E, et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010;20:1297–303. https://doi.org/10.1101/gr.107524.110
crossref pmid pmc
17. Purcell S, Neale B, Todd-Brown K, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 2007;81:559–75. https://doi.org/10.1086/519795
crossref pmid pmc
18. Zhou J, Liu L, Lopdell TJ, Garrick DJ, Shi Y. HandyCNV: standardized summary, annotation, comparison, and visualization of copy number variant, copy number variation region, and runs of homozygosity. Front Genet 2021;12:731355. https://doi.org/10.3389/fgene.2021.731355
crossref pmid pmc
19. Danecek P, Auton A, Abecasis G, et al. The variant call format and VCFtools. Bioinformatics 2011;27:2156–8. https://doi.org/10.1093/bioinformatics/btr330
crossref pmid pmc
20. Szpiech ZA, Hernandez RD. selscan: An efficient multithreaded program to perform EHH-based scans for positive selection. Mol Biol Evol 2014;31:2824–7. https://doi.org/10.1093/molbev/msu211
crossref pmid pmc
21. Gondro C, Porto-Neto LR, Lee SH. R for genome-wide association studies. Gondro C, van der Werf J, Hayes B, editorsGenome-wide association studies and genomic prediction. Methods in molecular biology. Humana Press; 2013. p. 1–17.
crossref
22. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 2010;26:841–2. https://doi.org/10.1093/bioinformatics/btq033
crossref pmid pmc
23. Wu T, Hu E, Xu S, et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2021;2:100141. https://doi.org/10.1016/j.xinn.2021.100141
crossref pmid pmc
24. Xu T, Guo X, Wang H, et al. Differential gene expression analysis between anagen and telogen of Capra hircus skin based on the de novo assembled transcriptome sequence. Gene 2013;520:30–8. https://doi.org/10.1016/j.gene.2013.01.068
crossref pmid
25. Diao X, Qin J, Dong C, He L, Zhang W. Integrative transcriptomic and metabolomic analyses reveal the role of melatonin in promoting secondary hair follicle development in cashmere goats. BMC Genomics 2025;26:200. https://doi.org/10.1186/s12864-025-11389-0
crossref pmid pmc
26. Ge W, Wang SH, Sun B, et al. Melatonin promotes cashmere goat (Capra hircus) secondary hair follicle growth: a view from integrated analysis of long non-coding and coding RNAs. Cell Cycle 2018;17:1255–67. https://doi.org/10.1080/15384101.2018.1471318
crossref pmid pmc
27. Ma S, Cao W, Ma X, et al. Metabolomics reveals metabolites associated with hair follicle cycle in cashmere goats. BMC Vet Res 2024;20:208. https://doi.org/10.1186/s12917-024-04057-0
crossref pmid pmc
28. Vasu M, Ahlawat S, Arora R, Sharma R. Deciphering the molecular drivers for cashmere/pashmina fiber production in goats: a comprehensive review. Mamm Genome 2025;36:162–82. https://doi.org/10.1007/s00335-025-10109-z
crossref pmid
29. Ceballos FC, Joshi PK, Clark DW, Ramsay M, Wilson JF. Runs of homozygosity: windows into population history and trait architecture. Nat Rev Genet 2018;19:220–34. https://doi.org/10.1038/nrg.2017.109
crossref pmid
30. Pemberton TJ, Absher D, Feldman MW, Myers RM, Rosenberg NA, Li JZ. Genomic patterns of homozygosity in worldwide human populations. Am J Hum Genet 2012;91:275–92. https://doi.org/10.1016/j.ajhg.2012.06.014
crossref pmid pmc
31. Joost S, Jacob T, Sun X, et al. Single-cell transcriptomics of traced epidermal and hair follicle stem cells reveals rapid adaptations during wound healing. Cell Rep 2018;25:585–97E7. https://doi.org/10.1016/j.celrep.2018.09.059
crossref pmid
32. Snippert HJ, Haegebarth A, Kasper M, et al. Lgr6 marks stem cells in the hair follicle that generate all cell lineages of the skin. Science 2010;327:1385–9. https://doi.org/10.1126/science.1184733
crossref pmid
33. Glotzer DJ, Zelzer E, Olsen BR. Impaired skin and hair follicle development in Runx2 deficient mice. Dev Biol 2008;315:459–73. https://doi.org/10.1016/j.ydbio.2008.01.005
crossref pmid pmc
34. Jia Q, Zhang S, Wang D, et al. Regulatory effects of FGF9 on dermal papilla cell proliferation in small-tailed Han sheep. Genes 2023;14:1106. https://doi.org/10.3390/genes14051106
crossref pmid pmc
35. Yan H, Jin M, Li Y, et al. miR-1 regulates differentiation and proliferation of goat hair follicle stem cells by targeting IGF1R and LEF1 genes. DNA Cell Biol 2022;41:190–201. https://doi.org/10.1089/dna.2021.0288
crossref pmid
36. Ma T, Li J, Jiang Q, Wu S, Jiang H, Zhang Q. Differential expression of miR-let7a in hair follicle cycle of Liaoning cashmere goats and identification of its targets. Funct Integr Genomics 2018;18:701–7. https://doi.org/10.1007/s10142-018-0616-x
crossref pmid
37. Brunner MAT, Jagannathan V, Waluk DP, et al. Novel insights into the pathways regulating the canine hair cycle and their deregulation in alopecia X. PLOS ONE 2017;12:e0186469. https://doi.org/10.1371/journal.pone.0186469
crossref pmid pmc
38. Hwang D, Lee H, Lee J, et al. Micro-current stimulation has potential effects of hair growth-promotion on human hair follicle-derived papilla cells and animal model. Int J Mol Sci 2021;22:4361. https://doi.org/10.3390/ijms22094361
crossref pmid pmc
39. Liang W, Chen X, Ni N, et al. Corticotropin-releasing hormone inhibits autophagy by suppressing PTEN to promote apoptosis in dermal papilla cells. Ann Med 2025;57:2490823. https://doi.org/10.1080/07853890.2025.2490823
crossref pmid pmc
40. Sohn KM, Jeong KH, Kim JE, Park YM, Kang H. Hair growth-promotion effects of different alternating current parameter settings are mediated by the activation of Wnt/β-catenin and MAPK pathway. Exp Dermatol 2015;24:958–63. https://doi.org/10.1111/exd.12827
crossref pmid


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

Copyright © 2025 by Asian-Australasian Association of Animal Production Societies.

Developed in M2PI

Close layer
prev next