- Research
- Open access
- Published:
Comparison of reproductive performance and functional analysis of spermatogenesis factors between domestic yak and semi-wild blood yak
BMC Genomics volume 26, Article number: 418 (2025)
Abstract
This study investigates differences in reproductive performance, testicular histology, and transcriptomic profiles between male Subei (SB; semi-wild) yaks and two domestic yaks, Gannan (GN) and Qinghai (QH). Key metrics including mating age, utilization time, breeding capacity, morphometric traits, and testicular indices were analyzed. SB yaks exhibited superior reproductive metrics, including earlier sexual maturity, prolonged utilization periods, and enhanced breeding capacity compared to GN and QH (P < 0.05). Morphologically, SB yaks demonstrated significantly greater body weight, and testicular dimensions. Compared with GN and QH yaks, the seminiferous tubules of SB yaks exhibited significantly larger spermatogenic cells and luminal cavities, along with a notably higher sperm density within the luminal cavity. Transcriptomic analysis identified 2,403 and 4,428 differentially expressed genes (DEGs) in GN vs. SB and QH vs. SB comparisons, respectively. Eight key genes (TPPP3, SMAD3, PAFAH1B3, BMP7, ARSA, CTNNB1, SMAD4, STAT3) and three pathways (Hippo, pluripotency regulation, TGF-β) were implicated in testicular development and spermatogenesis. These findings underscore the genetic and physiological advantages of SB yaks, offering insights for enhancing male yak reproductive performance.
The reproductive performance of male mammals is primarily reflected in testicular development, semen quality, and reproductive disorders. As a crucial indicator of fertilization capacity, semen quality significantly affects the conception rate in female animals [1]. The process of mammalian spermatogenesis is accomplished through precise coordinated regulation by numerous genes and hormones, among which genetic factors play a decisive role [2, 3]. The expression of these genes exhibits stage-specific characteristics with precise temporal and spatial regulation [4,5,6]. Numerous studies have demonstrated that spermatogenesis is a dynamic process involving the proliferation and differentiation of spermatogonial stem cells (SSCs) within the testes. The proper progression of this process directly determines semen quality and exerts a significant impact on the reproductive performance of male animals [7]. With the rapid development and widespread application of modern molecular biology technologies in the field of life sciences, investigating the regulatory mechanisms of spermatogenesis within yak testes at molecular levels, such as transcription and translation, has become one of the key research directions [8, 9]. Guowen Wang et al. revealed the key molecular mechanisms underlying the transition from gonocytes to spermatogonia and subsequent spermatogenesis by analyzing dynamic gene expression profiles in testes of domestic yaks across different age groups. They validated the functional roles of conserved genes such as CXCR4 in yaks, supporting their universal significance in mammalian sperm development [10]. Scholars have revealed key genes (Dmc1, Spo11, Tnp2, Crem) affecting male infertility in cattle-yak hybrids through comparative transcriptomic analyses of genomic differences among cattle-yak hybrids, yaks, and cattle, providing critical insights into yak reproductive physiology [11,12,13,14]. Through cross-species transcriptomics studies involving mice [15, 16], humans [17, 18], and yak [19], numerous genes influencing spermatogenesis have been identified. These discoveries not only deepen our understanding of fundamental biological mechanisms but also demonstrate significant application potential in medicine, agriculture, and ecological conservation.
Subei yaks, inhabiting the Yanchiwan National Nature Reserve, represent a unique genetic resource shaped by natural and artificial selection. While their superior growth and production traits are recognized, reproductive mechanisms remain underexplored. This study integrates histological and transcriptomic approaches to compare SB yaks with domestic GN and QH breeds, aiming to elucidate the molecular drivers of spermatogenesis in SB yaks, which holds significant research implications and value for yak breed improvement and the development of the industry.
Research methodology
Experimental animals and sample collection
The sexual maturity time, time of utilization, breeding ability, newborn weight, body weight, body height, oblique length of body, chest measurement, and body length of 4-year-old GN, QH, and SB yaks were compared by group investigation and access to records. The left testes of 4-year-old yaks were collected from the slaughterhouses of Qingyuan in Linxia, Huangyuan in Qinghai, Subei County in Jiuquan (10 yaks/copy), and the circumference of the testis, the long testis diameter, and the short testis diameter were measured and weighed. The tunica albuginea of the testes was incised with a scalpel, followed by epididymal removal. A transverse circumferential incision was made along the midsagittal plane of the testes to obtain the testicular parenchyma adjacent to the epididymal head. The parenchyma was dissected into 2 × 2 cm3 cubes and fixed in 4% paraformaldehyde for histological sectioning. Simultaneously, remaining tissue samples were flash-frozen in liquid nitrogen and transferred to a − 80 °C freezer for subsequent transcriptomic analysis and qRT-PCR validation.
H.E staining
Put the tissue sections on the dyeing rack in turn them into xylene I and xylene II in each 10 min, and in turn into different concentration gradient alcohol solutions for descending dewaxing hydration, distilled water washing 5 min, hematoxylin staining nucleus 5 ~ 6 min, distilled water rinsing 3 times (3 s/time), hydrochloric acid alcohol differentiation 2 ~ 3 s, into the tap water back to blue 10 min (running water), distilled water washing 3 min, eosin staining cytoplasm 3 min, distilled water rinsing 3 times (3 s/time), different concentration gradient alcohol solution ascending dehydration, xylene I, II in each 10 min, neutral resin glue sealing, airing at room temperature for 4 days.
RNA preparation and quality inspection
Samples of 100 mg each from the testicular tissues of GN, QH, and SB yaks were collected and finely ground in liquid nitrogen. The ground tissue was then transferred to a 1.5 mL centrifuge tube designed for non-RNA enzymes. Total RNA was extracted from the yak testicular tissue using the Trizol method. After extraction, the RNA was allowed to dry naturally at room temperature. An appropriate volume of DEPC-treated sterile water was added to dissolve the RNA completely, and the resulting solution was stored at − 80 °C in a freezer.
The purity and concentration of total RNA were detected by Nanodrop2000 ultramicro ultraviolet spectrophotometer. The samples with OD260/OD280 ≥ 1.8, OD260/230 ≥ 2.0 were qualified, and the total RNA was greater than 1 μg. The qualified total RNA can be used for subsequent experiments.
Library preparation and sequencing
RNA library construction was performed using the TruSeqTM RNA sample preparation Kit (Illumina, SanDiego, CA). Sequencing with the Illumina NovaSeq 6000 sequencer.
Raw data quality control and sequence alignment analysis
The raw data is quality controlled by fastp [20], and the low-quality data is filtered out to obtain clean reads. Then, the clean reads were compared with the yak reference genome sequence by TopHat2 and HISAT2 [21] application software to obtain mapped reads for expression calculation and subsequent transcript assembly.
Gene expression analysis
Stringtie [22] was used to reconstruct transcripts, and RSEM [23] was used to calculate the expression of all genes in each sample. Fragments Per Kilobase of transcript per Million mapped reads (FPKM) were used as a quantitative indicator.
Note: Let FPKM(A) be the expression of gene A, then C is the number of sequenced fragments compared to gene A, N is the total number of sequenced fragments compared to the reference gene, and L is the number of bases of gene A. The FPKM method eliminates the influence of the differences in gene lengths and sequencing quantities on the calculation of gene expression, and the calculated gene expression can be directly used to compare the differences in gene expression among different.
Screening of differentially expressed genes
After obtaining the number of gene read counts, the differential expression analysis of genes between different samples was performed by DESeq2 [24] application software, DEGs were identified, and then the function of DEGs was studied. In this study, |log2 (Fold Change)|≥ 2 and FDR < 0.05 were used as the screening conditions, and the genes that met this condition could be regarded as significant DEGs.
Gene Ontoiogy (GO) enrichment analysis of DEGs
In this study, Goatools application software was used to perform GO functional enrichment analysis on DEGs. After confirming the main GO functions of DEGs, Fisher was used for accurate test. In order to control the false positive of the calculation, the P-value was corrected by Benjamini and Hochberg multiple test method. According to the GO database, according to its different functions, it is divided into three aspects: molecular function (MF), cellular component (CC) and biological process (BP). When the P-value < 0.05, it is considered that this GO function is significantly enriched.
Target gene screening
Gene cards were used to screen out the genes related to testicular development and sperm production stores greater than 5 [25] and intersected with the DEGs related to reproduction screened by GO enrichment. PCA, PLSDA, and VIP analysis of variable projection importance were performed by MetaboAnalyst (https://www.metaboanalyst.ca/). VIP > 1 and P < 0.05 were used as criteria to further screen out the main genes related to spermatogenesis in GN, QH, and SB yaks.
Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of DEGs
KEGG enrichment analysis was performed on the target gene using the KOBAS online tool. Fisher was used for an accurate test, and the P-value was corrected by BH multiple tests. When the P-value < 0.05, it is considered that there is a significant enrichment of the KEGG function.
Regulation network analysis
High-confidence protein–protein interaction (PPI) networks were constructed via the STRING database (https://cn.string-db.org/) with a confidence threshold ≥ 0.7. Network topological parameters were analyzed using Cytoscape 3.7, and genes ranking in the top 10% for both Degree and Betweenness centrality values were selected. The resulting PPI network was visualized to identify key genes highly associated with spermatogenesis regulation.
Verification of DEGs
In order to further verify the reliability of RNA-Seq data, 11 DEGs were randomly selected for quantitative verification analysis by qRT-PCR. Quantitative primers were designed by Premier5.0 application software, and the yak β-actin gene was used as the internal reference gene. The specificity of the primers was detected by NCBI Primer Blast, and then sent to Shanghai Biological Biotechnology Co., Ltd. for synthesis. The primer information is shown in Table 1.
Data analysis
The relative quantitative method was used to calculate the transcription level of the gene by 2−△△Ct method with yak β-actin as the internal reference gene [26]. One-way ANOVA analysis of variance was performed using SPSS25.0 application software. Duncan's method was used for multiple comparisons when the difference was significant. The analysis results were expressed as Mean ± SEM. P < 0.05 indicated that the difference was significant, and P < 0.01 indicated that the difference was extremely significant.
Results and analysis
Comparative analysis of apparent performance between domestic yak and semi-wild blood yak
As shown in Table 2, the age at first mating of GN and QH yaks was significantly higher than that of SB yaks (P = 0.013). The service lifespan of SB yaks was highly significantly longer than those of GN and QH yaks (P < 0.01). Additionally, the breeding ability of SB yaks was highly significantly greater than that of QH yaks, while QH yaks exhibited a highly significant superiority in breeding capacity over GN yaks (P < 0.01).
Table 3 showed that the birth weight, body length, and body length of SB yaks were significantly higher than those of GN yaks, and the birth weight, body length, and body length of GN yaks were significantly higher than those of QH yaks (P < 0.01). The body weight of SB yaks were significantly higher than that of QH yaks (P = 0.028). Additionally, SB yaks displayed extreme dominance over GN yaks in withers height (P = 0.006) and highly significant superiority in chest girth relative to both GN and QH yaks (P < 0.01).
As delineated in Table 4, SB yaks and QH yaks exhibited highly significant increases in testicular circumference compared to GN yaks (P < 0.01). Specifically, SB yaks demonstrated superior testicular longitudinal diameter and testicular weight over QH yaks (P < 0.01), while QH yaks significantly surpassed GN yaks in these two metrics (P < 0.01). Furthermore, SB yaks displayed a highly significant advantage in testicular transverse diameter relative to both QH and GN yaks (P < 0.01).
Observation on testicular tissue morphology of domestic yak and semi-wild blood yak
The testicular tissue of yaks is organized into numerous seminiferous tubules, each composed of a basement membrane formed by myoid cells and interstitial cells, along with spermatogenic cells at varying developmental stages. Sertoli cells are interspersed among the spermatogenic cells, while the luminal space of these tubules contains spermatozoa exhibiting distinct morphological forms. As shown in Fig. 1(A-C), the seminiferous tubules of SB yaks exhibit a larger diameter compared to those of GN and QH yak breeds. Figure 1 (D-F) further demonstrates that spermatogenic cells in SB yaks display a more densely arranged cellular organization and larger cell morphology relative to GN and QH counterparts, and a significantly higher sperm density in the tubular lumen compared to domestic yaks.
Histomorphology observations on testis of domesticated yak and semi-wild blooded yak. Note: A, D: Histomorphology of the testis of the GN yak; B, E: Histomorphology of the testis of the QH yak; C, F: Histomorphology of the testis of the SB yak. SC: sertoli cell; LC: leydig cell; SP: spermatozoon; Sg: Spermatogenic cell. Bar = 200 μm (100x), Bar = 50 μm (400x)
Comparative analysis of testicular reference genome between domestic and semi-wild blood yak
According to Table 5, a total of 40,915,328 unique reads (89.27%) and 42,175,453 total reads (92.01%) were mapped to the reference genome in the GN1 data. A total of 34,221,850 unique reads (89.97%) and 35,400,235 total reads (93.07%) were mapped to the reference genome in the GN2 data. A total of 35,327,230 unique reads (89.17%) and 36,396,938 total reads (92.87%) were mapped to the reference genome in GN3 data. A total of 35,132,313 unique reads (88.23%) and 36,203,804 total reads (90.92%) were mapped to the reference genome in QH1 data. A total of 32,927,273 unique reads (89.76%) and 33,985,097 total reads (92.64%) were mapped to the reference genome in QH2 data. A total of 38,050,488 unique reads (89.11%) and 39,258,579 total reads (91.94%) were mapped to the reference genome in QH3 data. A total of 40,818,978 unique reads (89.77% %) and 42,161,107 total reads (92.72%) were mapped to the reference genome in SB1 data. A total of 39,892,436 unique reads (89.93%) and 41,180,952 total reads (92.83%) were mapped to the reference genome in SB2 data. A total of 31,725,343 unique reads (89.26%) and 32,784,445 total reads (92.24%) were mapped to the reference genome in SB3 data. It could be seen that the accuracy and coverage of the sequencing results were high and could be used for subsequent data analysis.
Distribution of gene expression between domestic and semi-wild blood yak testicular samples
The violin plot (Fig. 2) showed the distribution of gene expression between 9 testicular samples. The maximum, median, and minimum values of gene expression in 9 samples were similar, indicating that the reliability of gene expression results obtained by sequencing was high, and the gene expression levels between groups were similar.
Correlation analysis between domestic and semi-wild blood yak testicular samples
Based on the expression information, used R (http://www.r-project.org/) to carry out principal component analysis (PCA). As illustrated in Fig. 3(A), the PCA scatter plot of samples revealed strong intra-group reproducibility, with SB testicular samples clustering tightly together, indicating high similarity within the group. Distinct inter-group differentiation was also observed. Pearson correlation coefficients were calculated to quantify expression-level relationships between any two samples, visualized as a heatmap in Fig. 3(B). All samples exhibited robust correlations (> 95%), except for GN2, which showed lower correlation with other samples (≥ 63%). It may be due to the hybridization between GN yaks and domestic yaks in different regions. This phenomenon may be attributed to the lag in local domestication processes, or it could result from varying influences of altitude, climate, temperature, and hybridization on different individuals, which has led to the formation of class group within the same breed of domestic yaks in specific regions. However, no significant differences in morphological characteristics are observed among these domestic yak populations.
Correlation analysis of sample. Note: (A) The PC1 coordinate indicates the first principal component, while the percentage in parentheses indicates the contribution of the first principal component to the sample variance; the PC2 coordinate indicates the second principal component, while the percentage in parentheses indicates the contribution of the second principal component to the sample variance. B The coloured points in the graph indicate the individual samples respectively. Both the horizontal and vertical coordinates in the graph are for each sample, and the colour shade indicates the size of the correlation coefficient between the two samples. The closer the colour is to red, the higher the correlation, and the closer the colour is to blue, the lower the correlation
Basic analysis of differential genes in testis of domestic and semi-wild blood yak
In this study, raw counts were analyzed by edgeR [27] software. Based on certain standardized processing, FDR < 0.05 and |log2FC|≥ 2 were used as criteria to screen DEGs between domestic and semi-wild blood yak testis groups. From Fig. 4(A), comparative analysis of DEGs revealed distinct transcriptional profiles among yak populations. When comparing GN yaks versus SB yaks, we identified 2,403 DEGs (687 up-regulated; 1,716 down-regulated). QH yaks versus SB yaks, with 4,428 DEGs detected (1,493 up-regulated; 2,935 down-regulated). GN yaks versus QH yaks, with 4 genes showing differential expression (2 up-regulated and 2 down-regulated).According to Fig. 4(B), 1848 common DEGs were obtained among GN, QH, and SB yaks.
GO enrichment analysis of DEGs in the testis of domestic yak and semi-wild blood yak
A total of 1848 DEGs were enriched between GN, QH, and SB yak groups. In this experiment, Goatools application software was used to perform GO enrichment analysis on these DEGs, which were significantly enriched into 1736 GO terms, including 1314 biological processes, 192 cellular components, and 230 molecular functions.
Figure 5 showed the second-level histogram of GO functional enrichment results classification. We summarized the results of GO enrichment differences and removed duplicate genes to obtain 51 DEGs (STK4, NOTCH1, LRP2, TPPP3, TBX3, NIPBL, SIN3 A, SMAD3, USP47, ROS1, SRF, CTC1, ARID4 A, INPP5B, MAP2 K1, NKX3 - 1, TGFBR1, PUM1, CLOCK, PAFAH1B3, ZBTB16, HERC2, CXADR, NOTCH2, ASH1L, BRCA2, LHFPL2, ACSL4, STAT3, BMP7, CTNNB1, ARSA, B4GALT1, TUT4, CREBRF, PSAP, BMPR2, TERF1, SMC1 A, FANCM, KIT, DLG1, PSMD13, NDC1, SMALF1, HIFFM1, NRD4) highly associated with spermatogenesis. These genes were mainly enriched in Cell, Cell part, Organelle, Organelle part, Membrane, and Macromolecular complex, and were related to biological processes such as Binding, Catalytic activity, Molecular function regulator, Transporter activity, Signal transducer activity, Molecular transducer activity, Nucleic acid binding transcription factor activity, Transcription factor activity—protein binding, and Structural molecule activity. They were involved in the physiological processes of Cellular process, Single-organism process, Metabolic process, Biological regulation, Regulation of biological process, Response to stimulus, Cellular component organization or biogenesis, Positive regulation of biological process, Localization, Multicellular organismal process, Signaling, Negative regulation of biological process, Immune system process, Locomotion, Multi—organism process, Biological adhesion, Reproductive process, Reproduction, Growth, Behavior, and Rhythmic process in GN/QH and SB yaks. Among them, TPPP3, SMAD3, BMP7, PAFAH1B3, and ARSA exhibited up-regulated expression in SB yak testes.
Screening of target genes in domestic yaks and yaks of semi-wild origin
According to Fig. 6(A), PCA divided DEGs into two principal components, of which the PC1 contribution rate was 86.4%, the PC2 contribution rate was 9%, and the total contribution rate was 95.4%. And domestic yaks and semi-wild blood yaks have good discrimination. Figure 6(B) showed that the PLS-DA analysis results of 51 DEGs, and the contribution rate of PLS-DA model was 92.4%. Among them, the differential genes of domestic yak and semi-wild blood yak were obviously separated, indicating that the model had good classification prediction ability and stability. Figure 6(C) showed the screening of DEGs using the variable projection importance of PLS-DA, with VIP > 1 and P < 0.05 as the screening criteria. A total of 31 target genes (BMPR2, DLG1, KIT, ASH1L, INPP5B, BMPR1 A, SMC1 A, STK4, NSUN2, ACSL4, TGFBR1, LHFPL2, HERC2, SRF, STAT3, CREBRF, NIPBL, BMP7, TBX3, B4GALT1, USP47, HIF1 A, ARID4 A, PSMD13, PUM1, ARSA, CTC1, CTNNB1, FANCM, SMAD4, SMAD3) were screened out.
KEGG enrichment analysis of selected spermatogenesis-related genes
KEGG enrichment analysis was performed on the selected 31 target genes affecting yak spermatogenesis, and Fig. 7 showed the first 20 pathways with significant enrichment. The target genes were significantly enriched in the Hippo signaling pathway, Signaling pathways regulating the pluripotency of stem cells, and the TGF-beta signaling pathway.
Construction and visualization analysis of DEGs PPI network in the testis of domestic and semi-wild blood yak
Using 51 screened DEGs associated with reproduction, a PPI network was constructed based on network topological parameters (Fig. 8), which identified five core regulatory genes (CTNNB1, SMAD4, STAT3, SMAD3, NOTCH1) involved in spermatogenesis. Among them, CTNNB1, SMAD4, STAT3, SMAD3 and NOTCH1 had regulatory relationships with each other. CTNNB1 had a regulatory relationship with HIF1 A, TGFBR1, BMPR2, KIT, BMPR1 A, BRCA2, NOTCH2, MAP2 K1, TBX3 and CLOCK. SMAD4 had a regulatory relationship with HIF1 A, TGFBR1, BMPR2, KIT, BMPR1 A, BRCA2 and MAP2 K1. STAT3 had a regulatory relationship with HIF1 A, TGFBR1, BMPR2, KIT, NOTCH2, MAP2 K1 and ROS1. SMAD3 had a regulatory relationship with HIF1 A, TGFBR1, BMPR2, BMPR1 A and SRF. NOTCH1 had a regulatory relationship with HIF1 A, TGFBR1, KIT, NOTCH2, and SRF.
Analysis of DEGs expression levels affecting testicular development and spermatogenesis in domestic yaks and semi-wild yak
It could be seen from Table 6 that the expression levels of TPPP3, SMAD3, BMP7, PAFAH1B3, and ARSA in SB yaks testis were significantly higher than those in GN and QH yaks (P < 0.01). The expression levels of SMAD4, CTNNB1, and STAT3 in the testis of GN and QH yaks were significantly higher than those of SB yaks (P < 0.01). However, no significant difference was no significant difference in NOTCH1 expression (P = 0.327). Consequently, eight key genes (TPPP3, SMAD3, PAFAH1B3, BMP7, ARSA, CTNNB1, SMAD4, and STAT3) influencing SB yak spermatogenesis were identified.
Verification of DEGs in testis of domestic and semi-wild blood yak by qRT-PCR
Eleven DEGs (FGFR1, MAP2 K1, STAT3, PIK3 CA, ACVR2 A, HIF1 A, BMPR1 A, ROS1, zfhx3, PAK2, ROBO2) were selected, Yak β-actin was used as the internal reference gene, the same yak testis RNA sample as RNA-Seq sequencing was used for fluorescence quantitative PCR verification. The verified trend of gene expression was consistent with the sequencing results, which proved that the results of RNA-seq transcriptome data were reliable (Fig. 9).
Discussions
Spermatogenesis is a complex process that involves structural and biochemical changes in the seminiferous epithelium of adult animals and promotes the spread of genetic resources [28]. Mammalian spermatogenesis is regulated by paracrine, autocrine, and endocrine pathways as well as a series of structural elements and chemical factors that regulate the activity of somatic and germ cells [29,30,31]. There are many changes in gene expression in the process of spermatocyte to spermatogonia transformation [32]. The testicular development of 3 ~ 5 month-old yak is regulated by gene dynamic expression. Comparative analysis of DEGs reveals substantial transcriptional shifts during three critical phases of spermatogenic cell maturation, particularly marking the transition from spermatogonia to spermatocytes [10]. And the communication network orchestrating cellular activities during spermatogenesis exhibits a highly intricate architecture [33, 34]. A complex regulatory interplay exists between genotypic determinants and phenotypic expression [32]. Transcriptomics is essential for understanding the regulation of genes in animal growth and development [35]. In this study, we compared the apparent characteristics and histological differences between domestic yaks and semi—wild blood yaks through measurement, follow—up investigations, data record access, H.E staining, and transcriptomics analysis. We also clarified the key genes and important pathways that affect the spermatogenesis of SB yaks. This work can assist breeders in selecting candidate genes and has significant implications for enhancing the reproductive potential of yaks.
Comparison of apparent performance and histology between domestic yak and semi-wild blood yak
The reproductive potential of bulls depends on their mating ability and fertilization ability [36]. The early development of body weight and the time to reach sexual maturity directly affect the mating ability and fertilization ability of bulls. The results of this experiment showed that the body size and body weight of SB yak are significantly or extremely significantly higher than GN and QH yak. Although there is no significant difference in sexual maturity time among the three kinds of yaks, the first mating time of SB yak is significantly lower than GN and QH yak, and the time of utilization and mating ability are significantly or extremely significantly higher than GN and QH yak. Studies have shown that larger yaks enter puberty earlier than smaller yaks [37]. Datong yak is the first artificially cultivated hybrid of wild and domestic yak, showing excellent growth characteristics and production performance. The high growth, development, and production rate of Datong yak indicate that it is feasible to improve the traits of domestic yak by using wild yak resources, and the potential importance of using wild yak genetic resources for yak breeding is clarified [38], which is consistent with the growth characteristics of semi-wild blood yak in this experiment.
The size and function of the testis were crucial indicators for evaluating and breeding male animals, and they were highly heritable [39]. The apparent traits of SB testis in this experiment are significantly higher than GN and QH yak, and the testicular tissue morphology shows that the seminiferous tubules of SB yak testis are larger than GN and QH yak. Foote et al. founded that the testicular size and body weight of Sahiwal bulls increased in a curvilinear manner from 0 to 100 months of age. They observed a significant positive correlation between testicular development and body weight. After 100 months of age, the testicular size of Sahiwal bulls stabilized, while ejaculatory volume and sperm density increased significantly. However, there was no significant effect on sperm motility, plasma membrane integrity, or normal replacement rate [40]. T.A. Bongso, M.R., et al. studied the relationship between testis size and body weight in goats aged 3 to 28 months. The results indicated a significant positive correlation between testis size and body weight. Notably, testis size increased significantly between 6 and 7 months of age, accompanied by a sudden growth of seminiferous tubules, which promoted spermatogenesis [41]. The size of the testis would increase with the size of the body [42]. Research had shown that larger mammals require proportionately larger organs to fulfill their physiological needs. The size and weight of the testis were positively correlated with both the rate of testicular sperm production and sperm reserves [43]. Testicular size was strongly linked to daily sperm production, as the amount of sperm produced per unit of testicular volume remains constant, and the overall sperm output primarily depends on testicular size [36]. A large number of studies had shown that mammalian body size and body weight were significantly positively correlated with testicular development [44, 45]. The weight and size of the testis directly affected the discharge and quality of semen [46,47,48,49,50]. Observations of body size, body weight, age of utilization, testicular characteristics, and tissue morphology indicate that SB yak exhibits strong heterosis. However, the reproductive capacity of SB yaks was significantly lower than that of GN and QH yaks. This may be due to the fact that the testicles produce sperm while simultaneously generating substantial amounts of semen and hormones, resulting in a greater dilution effect [51].
Transcriptome difference analysis between domestic yak and semi-wild blood yak
Through transcriptome screening, eight key genes impacting spermatogenesis in SB yak were identified: TPPP3, SMAD3, PAFAH1B3, BMP7, ARSA, CTNNB1, SMAD4, and STAT3. TPPP3 belongs to the tubulin polymerization-promoting protein (TPPP) family. It promotes cell proliferation by influencing the replication and separation of centrosomes during mitosis [52]. Microtubules (MTs) are polymers composed of tubulin heterodimers. These structures are distributed throughout cells and play crucial roles in diverse cellular functions. They participate in the elongation and guidance of neurons within the growth cone, the maintenance of cell shape and intracellular transport, and the formation of the mitotic spindle, which is indispensable for chromosome separation during cell division [52,53,54]. TPPP3 binds to microtubules and promotes their bundle and network formation in a concentration-dependent manner, increasing the level of microtubule acetylation [55]. Different domains of TPPP3 protein promote microtubule elongation, and TPPP3 plays a role in developing a ciliated cell basal body array, providing a loose but limited microtubule network to cope with external loads [56]. In the absence of TPPP3 protein, in addition to cell cycle arrest and down-regulation of several cyclin proteins, HeLa cells also observed abnormalities in the metaphase and/or anaphase of mitosis, forming multipolar spindles [52]. This resulted in abnormal centrosome amplification, thereby increasing the frequency of abnormal mitosis and chromosome segregation errors [57, 58]. Studies have demonstrated that TPPP3 dynamically regulates β-catenin activity in the endometrial microenvironment during embryo implantation, coordinating cellular adhesion and signaling pathways critical for successful implantation [59], and the Wnt/β-catenin signaling pathway is involved in regulating the occurrence of mature sperm cells [60, 61]. β-catenin is an important effector of the Wnt/β-catenin signaling pathway and a regulator of cell adhesion [62]. Its role in testicular development may be more as a cytoplasmic adaptor protein [63]. It is also an important molecular link that integrates the signal events necessary for the development and maturation of germ cells after sertoli cell-germ cell adhesion and meiosis. The β-catenin complex of spermatogenic cells and the apical ectoplasmic specialization of the β-catenin complex on sertoli cells on the surface of ES triggers a signal cascade to regulate the development and maturation of spermatogenic cells at the late stage of meiosis [64].
The Smad protein serves as a classic intracellular mediator in the signal—transduction process of the TGF-β superfamily. The signal transduction initiated by ligands such as activin, TGF-β, and BMP, in combination with the regulated expression and differential utilization of specific Smad proteins, plays a vital role in the accurate regulation of the responses of testicular cells to the ligands of the TGF-β superfamily [65]. The Smad protein is predominantly located in the cytoplasm of meiotic germ cells, Sertoli cells, and Leydig cells, where it serves a vital function in testicular development and spermatogenesis [66]. Smad3 and Smad4 are key signal—transduction proteins within the TGF-β signaling pathway. Signaling from the TGF-β superfamily is essential for the determination of primordial germ cells, the preservation of the integrity of fetal testicular development, and the regulation of spermatogenesis in adult testes [67, 68]. Smad3 gets activated by ligands like TGF-β, activin, nodal, myostatin, and GDF3/9. Subsequently, it forms a complex with Smad4 and translocates into the nucleus to control the expression of target genes, thereby playing a significant part in testicular development, maturation, and the maintenance of its function [69, 70]. The activation of the SMAD3 and AKT signaling pathways serves as the crucial factor in sustaining the long—term proliferation and undifferentiated state of human sperm stem cells. Through the establishment of a well-defined culture system that incorporates specific growth factors and hydrogels, human sperm stem cells have been successfully amplified and managed to remain in an undifferentiated state for as long as 2 months [71]. SMAD4 acts as a key regulator in signal transduction within the TGF-β superfamily. It experiences a cycle of ubiquitination and deubiquitination, which is regulated by the ubiquitin ligase Ecto/TIF1γ and the deubiquitinase FAM/USP9x. This dynamic regulatory process controls the transcriptional response to TGF-β by promoting the assembly and dissociation of the Smad signaling complex [72]. Double deletion of Smad4 in Sertoli and Leydig cells results in fetal testicular tubular dysplasia and mild testicular dysplasia in adulthood. These manifestations include reduced testicular volume, partial seminiferous tubule dysplasia, and decreased sperm production [73]. The experimental results demonstrated that SMAD3 expression was upregulated in SB yak testicular tissues, whereas SMAD4 expression exhibited a downregulated profile. This observation suggests that SMAD3 might compensate for the diminished SMAD4 levels at the critical regulatory element of the Fshb promoter, potentially maintaining transcriptional regulation efficiency [74]. Bone morphogenetic proteins (BMPs), which are part of the TGF-β superfamily, play a vital role in male fertility as well as the development of fetal germ cells and somatic cells post-birth [75]. Among them, BMP7 serves as a key signaling molecule in testicular development and spermatogenesis. It selectively activates particular SMAD receptor proteins, namely SMAD1 and SMAD5, and participates in the regulation of testicular development and spermatogenesis [76], Additionally, BMP7 promotes the proliferation of Sertoli cells [77], and is involved in regulating the proliferation and differentiation of spermatogonia [78]. BMP8a is capable of enhancing the proliferation of germ cells via the SMAD2/3 signaling pathway and guiding the differentiation of germ cells through the SMAD1/5/8 signaling pathway [79]. When one allele of Bmp7 is removed, it exacerbates the phenotype of Bmp8a deletion mutants in spermatogenesis and the epididymis. This finding implies that BMP7 and BMP8 transmit signals in these two systems by using the same or similar receptors [80].
Platelet-activating factor (PAF) can influence sperm motility and acrosome function and has a role in the development of the animal reproductive system [81]. The mRNA of platelet-activating factor acetylhydrolase 1b (PAFAH1B) is found in different stages of mouse testicular development and spermatogenesis. These stages involve spermatogonia, spermatocytes, and early sperm cells during meiosis as well as the early stages of haploid germ cell development [82]. Research has shown that the PAFAH1B3 protein is located on the mitotic spindle of spermatocytes undergoing meiosis, which suggests that it plays a significant part in spermatogenesis [83, 84]. The expression level of the PAFAH1B3 gene would affect the process of testicular development and spermatogenesis [85], and knockout of the PAFAH1B3 gene would lead to spermatogenesis disorder and increased apoptosis of sperm cells [86]. Arylsulfatase A (ARSA) is a lysosomal enzyme found in the male reproductive system [87]. It is present in the lysosomes of testicular Sertoli cells as well as the acrosomes of sperm cells and spermatozoa [88]. Sulfogalactosylglycerolipid (SGG) is selectively present in male germ cells. Both SGG and ARSA have a direct affinity for the zona pellucida (ZP) and play important roles in spermatogenesis and sperm-egg interaction [89]. SGG serves as the physiological substrate of lysosomal ARSA in sertoli cells. In ARSA-knockout mice, SGG accumulates in sertoli cells, leading to lysosomal accumulation. This accumulation damages the function of sertoli cells, resulting in impaired spermatogenesis and near-zero in vitro spermatogenesis [90]. Studies have shown that ARSA can degrade SGG, prevent SGG-induced cytotoxicity, and produce palmitylpalmitoylglycerol (PPG). This PPG is used to synthesize SGG in a new generation of primary spermatocytes [91]. Signal transducers and activators of transcription 3 (STAT3) is a multifunctional transcription factor. It can either activate or inhibit the transcription of target genes, thus influencing cell proliferation, survival, and apoptosis [92, 93]. STAT3 is predominantly expressed in sertoli cells [94], where it regulates the differentiation of spermatogonial stem cells through modulation of the downstream target gene Ngn3 [95]. Notably, pharmacological inhibition of STAT3 phosphorylation has been shown to downregulate Zfp637 expression, thereby impairing the normal spermatogenesis process [96]. This study revealed that in the testicular tissues of SB yaks, genes such as TPPP3, SMAD3, PAFAH1B3, BMP7, and ARSA exhibited significant upregulation, while the expression levels of CTNNB1, SMAD4, and STAT3 were markedly downregulated. These gene expression characteristics were highly consistent with the histomorphological observations of SB yak testicular tissues, specifically manifested as enlarged luminal diameters of seminiferous tubules, tightly arranged spermatogenic cell layers with increased nuclear volumes, and a significantly higher sperm density in the tubular lumen compared to domestic yaks.
In summary, this experiment compared the apparent performance and testicular tissue morphological characteristics of GN, QH, and SB yaks. During this process, we elucidated the strong advantages exhibited by male SB yaks in terms of growth performance, reproductive performance, and testicular tissue morphology. To further explore the intrinsic mechanisms of SB yaks in reproduction, we employed transcriptome analysis and successfully screened out 8 dominant genes closely related to testicular development and spermatogenesis in SB yaks. Notably, these genes showed significant enrichment in the Hippo signaling pathway, the signaling pathway regulating stem cell pluripotency, and the TGF—beta signaling pathway. This discovery not only implies that these signaling pathways may play a crucial role in the testicular development and spermatogenesis of SB yaks, but also provides a new perspective for our understanding of the male yak's reproductive physiology. It helps us to further reveal the molecular mechanisms of yak reproductive regulation and provides a solid theoretical basis for the genetic improvement and enhancement of reproductive efficiency of yaks.
Ethics statement
All animal experiments were approved by the Ethics Committee of Gansu Agricultural University, 2024 (GSAU-Eth-VMC- 2024–009).
Data availability
Data is provided within the manuscript or supplementary information files. The sequencing data were deposited into NCBI’s Sequence Read Archive (SRA) (BioProject ID: PRJNA1194783).
References
Mohsen Ghaleb A-M, Muath QAG, Aiman AA. Ahmed RA-H, Abdulaziz HA-J, Mohammed Waleed A, Ramzi AA, Mohammed SA, Ayman AS: Effect of different concentrations of resveratrol on the quality and in vitro fertilizing ability of ram semen stored at 5 °C for up to 168 h. Theriogenology. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.theriogenology.2020.05.001.
Sharpe RM. Environmental/lifestyle effects on spermatogenesis. Philos Trans R Soc Lond B Biol Sci. 2010;1546:365. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00120-022-01951-z.
Zhang XJ, Liu ML, Jia MC. Regulation of gene expression during spermatogenesis at transcriptional level. Hereditas. 2011;33(12):1300–7. https://doiorg.publicaciones.saludcastillayleon.es/10.3724/sp.j.1005.2011.01300.
Ostermeier GC, Dix DJ, Miller D, Khatri P, Krawetz SA. Spermatozoal RNA profiles of normal fertile men. The Lancet. 2002. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0140-6736(02)09899-9.
Ostermeier GC, Dix DJ, Krawetz SA. A bioinformatic strategy to rapidly characterize cDNA libraries. Bioinformatics. 2002;18(7):949–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/18.7.949.
Reid R, Dix DJ, Miller D, Krawetz SA. Recovering filter-based microarray data for pathways analysis using a multipoint alignment strategy. Biotechniques. 2001;30(4):762. https://doiorg.publicaciones.saludcastillayleon.es/10.2144/01304st01.
Haiqi C, Evan M, Anubhav S, Anisha L, Jilong L, Daniel L, Xiaolu N, Jim H, Jingtao G, Bradley RC, et al. Dissecting mammalian spermatogenesis using spatial transcriptomics. Cell Rep. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2021.109915.
Rosengarten RD, Balaji S, Janez K, Gad S: The long noncoding RNA transcriptome of Dictyostelium discoideum development. G3 (Bethesda Md) 2017 7(2);387–398. https://doiorg.publicaciones.saludcastillayleon.es/10.1534/g3.116.037150
C., Soleilhavoup, G., Tsikis, V., Labas, G., Harichaux, P. L, Kohnke: Ram seminal plasma proteome and its impact on liquid preservation of spermatozoa. J Proteomics 2014, 109(1):245–260. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jprot.2014.07.007
Guowen W, Yongchang L, Qilin Y, Shuonan X, Shike M, Ruiqi Y, Ruina Z, Gong-Xue J, Deqiang A, Qi-En Y. Gene expression dynamics during the gonocyte to spermatogonia transition and spermatogenesis in the domestic yak. J Anim Sci Biotechnol. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-019-0360-7.
Wan RD, Gao X, Wang GW, Wu SX, Yang QL, Zhang YW, Yang QE. Identification of candidate genes related to hybrid sterility by genomic structural variation and transcriptome analyses in cattle-yak. J Dairy Sci. 2025;108(1):679–93. https://doiorg.publicaciones.saludcastillayleon.es/10.3168/jds.2024-24770.
Mengli C, Xingdong W, Shaoke G, Yandong K, Jie P, Xian G. F1 male sterility in Cattle-Yak examined through changes in testis tissue and transcriptome profiles. Animals. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ani12192711.
Shaokang Z, Tingting C, Xinmao L, Shiyi C, Jie W, Songjia L, Xianbo J. Identification of novel lncRNA and Differentially Expressed Genes (DEGs) of testicular tissues among Cattle, Yak, and Cattle-Yak associated with male infertility. Animals. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ani11082420.
Yiwen Z, Shixin W, Guo-Wen W, Rui-Dong W, Qi-En Y. Single-cell analysis identifies critical regulators of spermatogonial development and differentiation in cattle-yak bulls. J Dairy Sci. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.3168/jds.2023-24442.
Adrienne Niederriter S, Xianing Z, Sarah M, Qianyi M, Gabriel LM, Christopher DG, Meena S, Kyle EO, Jun ZL, Saher Sue H. Single-Cell RNA sequencing of human, macaque, and mouse testes uncovers conserved and divergent features of mammalian spermatogenesis. Dev Cell. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.devcel.2020.05.010.
Kira LM, Daniel JS, Jamie M, Günter PW, Bluma JL: Evolutionary innovations in germline biology of placental mammals revealed by transcriptomics of first wave spermatogenesis in opossum. bioRxiv (Cold Spring Harbor Laboratory) 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/2023.06.17.545442
Κρινιώ Γ, John TL, John PS, Amir H, Benjamin DH, Haocheng Y, James FA, Aditya B. Unraveling the testicular seminoma tumor microenvironment by single-cell RNA-sequencing. J Clin Oncol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/jco.2024.42.4_suppl.522.
Mei W, Xixi L, Gang C, Yidong C, Geng A, Liying Y, Shuai G, Yanwen X, Yueli C, Ji D, et al. Single-Cell RNA Sequencing Analysis Reveals Sequential Cell Fate Transition during Human Spermatogenesis. Cell Stem Cell. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.stem.2018.08.007.
TserangDonko M, Xuemei C, Wangsheng Z, Peng Z, Zhixin C, Binglin Y, Haiwei L, Jikun W, Haibo W, Zhijuan W, et al. Single-cell transcriptome analysis and in vitro differentiation of testicular cells reveal novel insights into male sterility of the interspecific hybrid cattle-yak. BMC Genomics. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-023-09251-2.
Shifu C, Yanqing Z, Yaru C, Jia G. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/bty560.
Daehwan K, Ben L, Steven LS. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nmeth.3317.
Mihaela P, Geo P, Corina A, Tsung Cheng C, Joshua TM, Steven LS. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nbt.3122.
Bo L, Colin ND. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2105-12-323.
Michael IL, Wolfgang H, Simon A. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13059-014-0550-8.
Li S, Jin L, Guangfei W, Cheng W, Zhiping L. Molecular Mechanism of YuPingFeng in the Treatment of Asthma Based on Network Pharmacology and Molecular Docking Technology. Comput Math Methods Med. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2022/7364126.
Schmittgen TD, Livak KJ: Schmittgen TD, Livak KJAnalyzing real-time PCR data by the comparative C(T) method. Nat Protocols 3(6): 1101–1108. Nature Protocol 2008, 3(6):1101–1108. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nprot.2008.73
Mark DR, Davis JM, Gordon KS. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2009. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/btp616.
Antoine R, Bernard J, Charles P. Testicular development and spermatogenesis: harvesting the postgenomics bounty. Adv Exp Med Biol. 2009. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-0-387-09597-4_2.
Bernard J. The Sertoli-Germ Cell Communication Network in Mammals. Int Rev Cytol. 1993. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0074-7696(08)60766-4.
Jégou B, Sharpe R. 8 – Paracrine mechanisms in testicular control. Mol Biol Male Reprod Syst 1993. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/B978-0-08-091764-1.50012-X.
William HW. Regulation of mammalian spermatogenesis by miRNAs. Semin Cell Dev Biol. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcdb.2021.05.009.
Haiyue X, Wu S, Shengwei P, Wanhong L, Fadi L, Xiumei Y. Identification of key genes related to postnatal testicular development based on transcriptomic data of testis in Hu Sheep. Front Genet. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2021.773695.
Lucio G. Gonadal peptides as mediators of development and functional control of the testis: an integrated system with hormones and local environment. Endocr Rev. 1997. https://doiorg.publicaciones.saludcastillayleon.es/10.1210/er.18.4.541.
Jégou B, Pineau C, Dupaix A: Paracrine Control of Testis Function. Springer US 1970. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-0-585-38145-9_3
Prashanth S, Lisette JAK, Haja NK. Multi-omic data integration and analysis using systems genomics approaches: methods and applications in animal production, health and welfare. Genet Sel Evol. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12711-016-0217-x.
Parkinson TJ. Evaluation of fertility and infertility in natural service bulls. Vet J. 2004. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tvjl.2003.10.017.
Jigme W, Kesang W. Productive and reproductive traits of yak types in Bhutan: characteristics and comparisons. J Appl Anim Res. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/09712119.2017.1416616.
Wen Q, Shuang L, Nan W, Zhouxuan W, Jun X, Hui M, Shou-Dong Z. Main factors influencing the gut microbiota of Datong Yaks in mixed group. Animals. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ani12141777.
John PK. Understanding and evaluating bovine testes. Theriogenology. 2014. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.theriogenology.2013.09.001.
Ejaz A, Nasim A, Zahid N, Aleem M, Muhammad Sarwar K, Ashiq M, Muhammad Y. Relationship of age to body weight, scrotal circumference, testicular ultrasonograms, and semen quality in Sahiwal bulls. Trop Anim Health Prod. 2010. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11250-010-9668-1.
Bongso TA, Jainudeen MR, Zahrah AS. Relationship of scrotal circumference to age, body weight and onset of spermatogenesis in goats. Theriogenology. 1982;18(5):513–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0093-691X(82)90184-4.
Yanara A, Harold F, Suitberto Cabrera G, Yaritza A: Relationship between body and testicular measurements in young buffalo bulls in Cuba. Revista Colombiana De Ciencias Pecuarias 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.17533/udea.rccp.v30n2a05
Anders Pape M. Ejaculate quality, testes size and sperm production in mammals. Funct Ecol. 1989. https://doiorg.publicaciones.saludcastillayleon.es/10.2307/2389679.
Alade NK, Ezeokoli CL, Muhammed ID. The relationships between body weight, testicular weight, age and the scrotal circumference of goats in the semi-arid Maiduguri. Nigeria Animal Production Research Advances. 2010. https://doiorg.publicaciones.saludcastillayleon.es/10.4314/apra.v5i1.49820.
Almquist JO, Amann RP: Reproductive Capacity of Dairy Bulls. II. Gonadal and extra-gonadal sperm reserves as determined by direct counts and depletion trials; dimensions and weight of genitalia. J Dairy Sci 1961. https://doiorg.publicaciones.saludcastillayleon.es/10.3168/jds.s0022-0302(61)89939-6
Almquist JO, Branas RJ, Barber KA. Postpuberal changes in semen production of charolais bulls ejaculated at high frequency and the relation between testicular measurements and sperm output. J Anim Sci. 1976. https://doiorg.publicaciones.saludcastillayleon.es/10.2527/jas1976.423670x.
Foote R. Factors influencing the quantity and quality of semen harvested from bulls, rams, boars and stallions. 1978. https://doiorg.publicaciones.saludcastillayleon.es/10.2527/1978.47Supplement_II1x.
Amann RP. A Critical review of methods for evaluation of spermatogenesis from seminal characteristics. Andrology. 1981. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/j.1939-4640.1981.tb00595.x.
Yusuf A, Igbokwe IO. Testicular and related size evaluations in Nigerian Sahel goats with optimal cauda epididymal sperm reserve. Vet Med Intern. 2015. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2015/357519.
Smith M, David LM, Max SA, Parish NR, John DW, Wiltbank JΝ. Relationships among fertility, scrotal circumference, seminal quality, and libido in Santa Gertrudis bulls. Theriogenology. 1981. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0093-691x(81)90072-8.
Paul B. Sperm wars sperm competition and the evolution of animal mating systems Robert L. Smith BioScience. 1986. https://doiorg.publicaciones.saludcastillayleon.es/10.2307/1310399.
Wenbai Z, Xuanchun W, Lianxi L, Xiaoqi F, Zhihong Y, Weiwei Z, Renming H. Depletion of tubulin polymerization promoting protein family member 3 suppresses HeLa cell proliferation. Mol Cell Biochem. 2009. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11010-009-0208-0.
Emma H, János K, Judit O, Ferenc O, Katalin FM, Judit O. Brain-specific p25 protein binds to tubulin and microtubules and induces aberrant microtubule assemblies at substoichiometric concentrations. Biochemistry. 2002. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/bi020140.
László T, Emma H, Judit O, István H, Ferenc O, Bálint S, János K, János S, Judit O. TPPP/p25 promotes tubulin assemblies and blocks mitotic spindle formation. Proc Natl Acad Sci USA. 2003. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.2436331100.
Judit O, Tibor S, Sándor S, Adél S, Attila L, Judit O. Further evidence for microtubule-independent dimerization of TPPP/p25. Sci Rep. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep40594.
Takayuki T, Shuji I, Kazuhiro O. Tubulin Polymerization-Promoting Protein Family Member 3 (Tppp3) Facilitates Microtubule Bundling and Network Formation via its Weak Interaction with Microtubules. Biophys J. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bpj.2018.11.1399.
William SS. Centrosomal amplification and spindle multipolarity in cancer cells. Semin Cancer Biol. 2005. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcancer.2004.09.003.
Kenji K, Izumi H, Zhenqiang M, Ryosuke I, Manabu M, Tatsuro T, Takayuki N, Linda L, Kohzaburo F-Y, Koji S et al: Data from induction of centrosome amplification and chromosome instability in human bladder cancer cells by p53 mutation and cyclin e overexpression. null 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.c.6493997
Vinay S, Pooja P, Jyoti K, Kanchan G, Anila D. Uterine TPPP3 plays important role in embryo implantation via modulation of β-catenin†. Biol Reprod. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/biolre/ioy136.
Jon MO, Ralph LB. The germline stem cell niche unit in mammalian testes. Physiol Rev. 2012. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/physrev.00025.2011.
Hans C, Kyle ML, Roel N. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science. 2014. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.1248012.
Andrew JA, Alexis F, David MP: Wnt/β-catenin regulates an ancient signaling network during zebrafish scale development. eLife 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/elife.37001
Maria Lucia C, Francesca F, Katia Corano S, Gabriella D, Fabio C, Paola G, Angela C, Giulia R. R-Spondin 1/Dickkopf-1/Beta-Catenin machinery is involved in testicular embryonic angiogenesis. PLoS ONE. 2015. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0124213.
Yao FuC. Jennifer SL-C, Krystle YH, Amiya PSH, Manjeet KR: Role of β-Catenin in post-meiotic male germ cell differentiation. PLoS ONE. 2011. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0028039.
Catherine I, Kate L. Smads and cell fate: Distinct roles in specification, development, and tumorigenesis in the testis. IUBMB Life. 2013. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/iub.1115.
Jian X, Amanda B, William HW, Elizabeth AM. Developmental and stage-specific expression of Smad2 and Smad3 in rat testis. Andrology. 2003. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/j.1939-4640.2003.tb02662.x.
Daniel JB. SMAD Expression in the testis predicts age- and cell-specific responses to activin and TGFβ. Andrology. 2003. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/j.1939-4640.2003.tb02663.x.
Catherine I, Sirisha M, Badia Maria B, Kate L. All in the family: TGF-β family action in testis development. Reproduction. 2006. https://doiorg.publicaciones.saludcastillayleon.es/10.1530/rep.1.01075.
Keijo L, Antti Y, Tomi PM. Developmentally regulated expression of Smad3, Smad4, Smad6, and Smad7 involved in TGF-beta signaling. Mech Dev. 2001. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0925-4773(00)00556-6.
Kate L, Dias VL, Sarah JM, Ewa Rajpert-De M. The transforming growth factor-β superfamily in early spermatogenesis: potential relevance to testicular dysgenesis. Andrology. 2007. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1365-2605.2007.00785.x.
Ying G, Linhong L, Min S, Yanan H, Zheng L, Zuping H. Expansion and long-term culture of human spermatogonial stem cells via the activation of SMAD3 and AKT pathways. Exp Biol Med. 2015. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1535370215590822.
Jeffrey LW. The Secret Life of Smad4. Cell. 2009. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2008.12.028.
Denise RA, Hisayuki Y. Loss of Smad4 in sertoli and leydig cells leads to testicular dysgenesis and hemorrhagic tumor formation in mice1. Biol Reprod. 2014. https://doiorg.publicaciones.saludcastillayleon.es/10.1095/biolreprod.113.111393.
Yining L, Gauthier S, Ulrich B, Chu Xia D, Jonathan MG, Daniel JB. SMAD3 regulates follicle-stimulating hormone synthesis by pituitary gonadotrope cells in vivo. J Biol Chem. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.m116.759167.
Catherine I, Kate L. SMAD expression in the testis: An insight into BMP regulation of spermatogenesis. Dev Dyn. 2007. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/dvdy.21401.
He-He H, Dan-Qian C, Yanni W, Ya-Long F, Gang C, Nosratola DV, Ying-Yong Z. New insights into TGF-β/Smad signaling in tissue fibrosis. Chem Biol Interact. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cbi.2018.07.008.
Manuela P, Paola G, Pellegrino R, Raffaele G, Susanna D. Developmental expression of BMP4/ALK3/SMAD5 signaling pathway in the mouse testis: a potential role of BMP4 in spermatogonia differentiation. J Cell Sci. 2003. https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jcs.00650.
Rossella P, Micaela M, Pieranna C, Mario S, Carla B. Regulatory role of BMP2 and BMP7 in spermatogonia and Sertoli cell proliferation in the immature mouse. Eur J Endocrinol. 2004. https://doiorg.publicaciones.saludcastillayleon.es/10.1530/eje.0.1510511.
Fang W, Ting-Yu L, Li Ying S, Wen-Liang C, Po Chih W, Chwan Yau L. BMP8A sustains spermatogenesis by activating both SMAD1/5/8 and SMAD2/3 in spermatogonia. Science Signaling. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/scisignal.aal1910.
Guang-Quan Z, Ya-Xiong C, Xiaoming L, Xu Z, Xiaoxia Q. Mutation in Bmp7 exacerbates the phenotype of Bmp8a mutants in spermatogenesis and epididymis. Dev Biol. 2001. https://doiorg.publicaciones.saludcastillayleon.es/10.1006/dbio.2001.0448.
Hiroyuki K, Noritaka Y, Mitsuharu H, Tomo-o I, Junken A, Makoto Mark T, Kazuhide I, Hiroyuki A. Targeted Disruption of intracellular type I platelet activating factor-acetylhydrolase catalytic subunits causes severe impairment in spermatogenesis. J Biol Chem. 2003. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.m211836200.
Wei Y, Amir HA, Anthony WB, Gregor E, Martin MM, Gary DC. Previously uncharacterized roles of platelet-activating factor acetylhydrolase 1b complex in mouse spermatogenesis. Proc Natl Acad Sci USA. 2003. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1236145100.
Lynn V, Björn H, Katrien S, Katarzyna S, Nerea OE, Minerva F-B, Krishna Chaitanya P, Luc P, Dieter D, Petra De S et al: Platelet-activating factor acetylhydrolase 1B3 (PAFAH1B3) is required for the formation of the meiotic spindle during in vitro oocyte maturation. Reproduction, Fertility and Development 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1071/rd18019
Nadja D: The role of Lissencephaly-1 protein in male germ cell differentiation. null 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.53846/goediss-3636
Scott BT, Nels CO, George LL, Edwin GB. Novel isoforms of intracellular platelet activating factor acetylhydrolase (PAFAH1b2) in human testis; encoded by alternatively spliced mRNAs. Prostaglandins Other Lipid Mediat. 2008. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.prostaglandins.2007.10.005.
Britta W, Ulrich S. Stem cell fate regulation by dynein motor protein Lis1. Nat Genet. 2014. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ng.2902.
Eduard T, Paul FC, Guangping C. para-Nitrophenyl Sulfate Activation of Human Sulfotransferase 1A1 Is Consistent with Intercepting the E·PAP Complex and Reformation of E·PAPS. J Biol Chem. 2009. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.m109.049312.
Wattana W. Acquisition of arylsulfatase a onto the mouse sperm surface during epididymal transit. Biol Reprod. 2003. https://doiorg.publicaciones.saludcastillayleon.es/10.1095/biolreprod.102.010231.
Eurı́dice C, Wattana W, Hongbin X, Arvan LF, Araya A, Ali S, Krittalak C, Nongnuj T: Binding of arylsulfatase a to mouse sperm inhibits gamete interaction and induces the acrosome reaction1. Biol Reprod 2002. https://doiorg.publicaciones.saludcastillayleon.es/10.1095/biolreprod66.6.1820
Kessiri K, Arpornrad S, Wongsakorn K, Mark AB, Kym FF, Dylan B, Nongnuj T. Accumulation of seminolipid in sertoli cells is associated with increased levels of reactive oxygen species and male subfertility: studies in aging arsa null male mice. Antioxidants. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antiox10060912.
Hongbin X, Kessiri K, Suraj K, Charles ES, Tony R, Naoko GI, Louis H, Kym FF, Nongnuj T. Arylsulfatase A deficiency causes seminolipid accumulation and a lysosomal storage disorder in Sertoli cells. J Lipid Res. 2011. https://doiorg.publicaciones.saludcastillayleon.es/10.1194/jlr.m019661.
Zhaojuan Y, Bo X, Xiaopeng H, Xiaoying Y, Yiqun T, Cuifeng Q, Shuzeng W, Haifeng C, Xiaohui B, Ji W. Dynein axonemal intermediate chain 2 plays a role in gametogenesis by activation of Stat3. J Cell Mol Med. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jcmm.13945.
Yuan M, Guizhen H, Jun L, Jie Z, Fēi L, Kai L, Bo G, Li Z, Wanying S, Ping L, et al. Hyperleptinemia directly affects testicular maturation at different sexual stages in mice, and suppressor of cytokine signaling 3 is involved in this process. Reprod Biol Endocrinol. 2014. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1477-7827-12-15.
Keiya N, Kasane I-K, Aya U, Ryuji H, Masamichi K, Masami KA. Regionally distinct patterns of STAT3 phosphorylation in the seminiferous epithelia of mouse testes. Mol Reprod Dev. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/mrd.22962.
Hairui T, Wang X, Xiaoxiao L, Weixiang S, Jiaqi M, Kang Z. Regulation of spermatogonial stem cell differentiation by Sertoli cells-derived exosomes through paracrine and autocrine signaling. J Cell Physiol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcp.31202.
Guizhen H, Yuan M, Jie Z, Jun L, Di G, Yanyan L, Ping L, Lei H. IL-6 mediates differentiation disorder during spermatogenesis in obesity-associated inflammation by affecting the expression of Zfp637 through the SOCS3/STAT3 pathway. Sci Rep. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep28012.
Acknowledgements
Thank the National Natural Science Foundation of China (NO.32273091, NO.32473110) for supporting this experiment. This experiment was supported by Jiuquan Animal Husbandry and Veterinary General Station and Subei Mongolian Autonomous County Animal Husbandry and Veterinary Technical Service Center.
Funding
This work was supported by the Natural Science Foundation of China (NO.32273091, NO.32473110).
Author information
Authors and Affiliations
Contributions
Conceptualization: S Y, Y C, Y P, and X P. Methodology: X P, Y P, and M W. Software: X P, S Q, and Y P. Validation: X P, and S Q. Resources: T Y and Y H. Data curation-Writing-Original draft preparation: X P. Supervision: S Y, Y C, and Y P. Project administration: Y P and S Y. Funding acquisition: S Y. Promotion of yak genetic resources in Subei: Y H, Y R, and T Y. All authors have read and agreed to the published version of the manuscript.
Corresponding authors
Ethics declarations
Consent for publication
All authors have read and agreed to the published version of the manuscript.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Pang, X., Pan, Y., Wang, M. et al. Comparison of reproductive performance and functional analysis of spermatogenesis factors between domestic yak and semi-wild blood yak. BMC Genomics 26, 418 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-025-11594-x
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-025-11594-x