Skip to main content

Epigenetic regulation of functional candidate genes for milk production traits in dairy sheep subjected to protein restriction in the prepubertal stage

Abstract

Background

As the prepubertal stage is a crucial point for the proper development of the mammary gland and milk production, this study aims to evaluate how protein restriction at this stage can affect methylation marks in milk somatic cells. Here, 28 Assaf ewes were subjected to 42.3% nutritional protein restriction (14 animals, NPR) or fed standard diets (14 animals, C) during the prepubertal stage. During the second lactation, the milk somatic cells of these ewes were sampled, and the extracted DNA was subjected to whole-genome bisulfite sequencing.

Results

A total of 1154 differentially methylated regions (DMRs) were identified between the NPR and C groups. Indeed, the results of functional enrichment analyses of the genes harboring these DMRs suggested their relevant effects on the development of the mammary gland and lipid metabolism in sheep. The additional analysis of the correlations of the mean methylation levels within these DMRs with fat, protein, and dry extract percentages in the milk and milk somatic cell counts suggested associations between several DMRs and milk production traits. However, there were no phenotypic differences in these traits between the NPR and C groups.

Conclusion

In light of the above, the results obtained in the current study might suggest potential candidate genes for the regulation of milk production traits in the sheep mammary gland. Further studies focusing on elucidating the genetic mechanisms affected by the identified DMRs may help to better understand the biological mechanisms modified in the mammary gland of dairy sheep as a response to nutritional challenges and their potential effects on milk production.

Peer Review reports

Introduction

The world population is constantly increasing, and the population size is expected to reach 10.9 billion by 2100 [1]. Consequently, a demand for increased production of food with high nutritional value is expected to emerge. However, sustainability and animal welfare standards must be maintained with this increase in production, which is a challenge for producers [2]. In animal production systems, management decisions related to feeding strategies can account for up to 75% of all variable costs in a herd, and protein accounts for a high proportion of these costs [3,4,5]. Several additional issues are associated with protein intake in livestock species. For example, in Europe, a disturbance of the nutrient cycle related to the supply of protein for animal feed is observed due to geographical separation, mainly concerning the import of soybeans from subtropical regions [6]. Consequently, the protein intake in flocks must be reviewed as a crucial component in controlling the sustainability of the whole production chain. Equally important, protein prices and availability in the market have recently been highly volatile. Therefore, feeding strategies in livestock herds are under constant pressure to change and adapt.

In general, nutrition is a major component affecting sheep milk composition, and appropriate nutritional management is directly related to improvements in the nutritional value of the milk [7]. In ruminants, postnatal mammary growth occurs at an allometric rate before puberty and returns to an isometric rate after puberty [8]. In general, sheep reach puberty at 6–8 months old, and there is a consensus that the ovine mammary allometric growth occurs between 3 and 4 months of age [9, 10]. It is well documented that elevated nutrient intake during this allometric growth phase results in reduced parenchymal mass and DNA [11]. The amplification of genomic DNA during lactation is hypothesized to be associated with increasing gene copies to support a high rate of RNA and protein synthesis [12]. Therefore, the nutrient supply during the critical developmental window and the diet composition are important factors affecting proper animal development and production to obtain high-quality products that meet consumer demands. Generally, farmers avoid reducing nutrition costs during lactation, trying to maintain the ewe’s productive potential. On the other hand, the effects of restricted feeding (reduction in concentrate) during the prepubertal stage of Dorset ewes were shown to improve mammary gland development without affecting growth performance [13]. In addition, in dairy cows, supplementation with protein during the prepubertal stage did not seem to affect milk production later in the lives of these animals [14, 15]. Interestingly, a previous study by our group revealed the absence of an effect of a 42.3% nutritional protein restriction (NPR) in the prepubertal growth stage on economically important traits in Spanish Assaf ewe lambs, such as milk somatic cell counts (SCC) [16]. This reinforces the notion that reducing protein intake in the diet of replacement ewe lambs may be performed without negatively impacting their milk production as adult ewes. The absence of phenotypic differences between groups of animals fed under regimes with substantial differences in protein intake during prepuberty might be due to compensatory biological mechanisms. Indeed, among the potential biological alterations associated with NPR, DNA methylation stand out as relevant candidates due to previously reported effects caused by protein restriction [17, 18]. Modifications in nutritional status can directly affect enzymes related to epigenetic processes or even change the availability of substrates for those enzymes [19]. Consequently, these alterations might affect the later expression of genes related to alterations in the mammary gland of ewes subjected to NPR in the prepubertal stage.

In light of the above, the main objectives of this study were 1) to evaluate the impact on the genome methylation state of the milk somatic cells during the second lactation of Assaf ewes subjected to NPR during the prepubertal stage and 2) to identify DNA methylation in functional candidate genes that are associated with regulatory mechanisms related to milk production traits in dairy sheep.

Material and methods

Sampling and nutritional challenge

Initially, 40 lamb ewes from a single flock in the northwest region of Castilla y León (Spain) that were transported to the facilities of the IGM in León were fed ad libitum with a standard diet for replacement ewe lambs providing 16% crude protein until three months of age and were subsequently divided into two groups. The two experimental groups were composed of 20 NPR and 20 C animals. To evaluate the impact of the protein restriction challenge due to a trade market problem and a shortage of concentrate inputs, the C ewes received the standard diet mentioned above for 64 d; during the same period, the NPR ewes received the same diet but replacing soybean meal with maize and barley grains. The diet composition offered to each group is shown in Supplementary Table 1. The NPR diet was planned to produce an intense reduction in dietary crude protein (42.3%) but has other notable qualitative changes, such as a 16% reduction in acid detergent fiber (ADF), a nearly 29% increase in acid detergent lignin (ADL) and ether extract. The diets were offered ad libitum for both groups. The 64-d NPR period in the prepubertal stage was coincident with the allometric growth of the mammary gland [11]. After the experimental period, the animals were fed ad libitum with the standard diet for replacement ewe lambs till milk sampling. The ewes passed through two gestational and lactation periods, where in the second lactation, the milk samples were collected for DNA extraction. Only 28 (out of the initial 40 challenged ewes) were pregnant in the second lactation. Therefore, these ewes constituted the sample used for the methylation marks analysis.

DNA extraction and whole-genome bisulfite sequencing

The milk samples (100 mL, 50 from each teat) obtained from 28 animals out of the 40 initially used in the nutritional challenge were collected once before the morning milking. Milk somatic cells were obtained using the following protocol: 1) addition of 50 µL of EDTA 0.5 mol/L to each sample; 2) centrifugation at 6000 × g for 10 min at 4 °C; 3) transfer of the pellet to a 2 µL microtube and centrifugation at 7000 g for 10 min at 4 °C; 4) removal of the supernatant and addition of 1 mL of wash buffer (15 mmol/L Tris–HCl (pH 7.4–7.6), 25 mmol/L NaCl, 5 mmol/L MgCl2, 15 mmol/L Na2HPO4, 2.5 mmol/L EDTA, 1% sucrose); 5) centrifugation at 3000 g for 3 min at 4 °C; 6) removal of the supernatant and dissolution of the pellet in 1 mL of wash buffer; 7) centrifugation at 3,000 g for 3 min at 4 °C (the last two steps can be repeated if the supernatant is not clear); finally, removal of the supernatant and DNA extraction. DNA extraction was performed using the MasterPure™ Complete DNA and RNA Purification Kit. The SCC and the DNA concentration (ng/mL) obtained for each sample are available on Supplementary Fig. 1. The samples were used for paired-end (150 bp) library construction on the Novogene platform (Milton, Cambridge, United Kingdom), and libraries were sequenced on an Illumina NovaSeq 6000 system, with a minimum coverage depth of 20X for each sample. Detailed information regarding library preparation and whole genome bisulfite sequencing (WGBS) is available from Fonseca et al. (2022) [20]. The raw datasets derived from sequencing are available in the European Nucleotide Archive (ENA) repository under accession number PRJEB56589 (https://www.ebi.ac.uk/ena/browser/view/PRJEB56589).

Differential methylation between nutritionally challenged and control sheep

The quality control of the 28 raw samples obtained from the aforementioned milk samples was performed using FastQC [21]. Subsequently, the reads were trimmed based on quality scores, adapters were removed, and short reads were filtered using the default options of Trim Galore software (version 0.6.5) [22]. Initially, the ovine reference genome, Oar_Ram_v2.0, was indexed by BowTie2 [23]. Subsequently, the Python script bs_seeker2-align.py from Bsseeker2 [24] was used to align the trimmed reads against the reference genome with the default options. The alignment output files were sorted by position using SAMtools software (version 1.15.1) [25], and duplicate reads were removed using Picard software (version 2.25) (https://broadinstitute.github.io/picard/). Finally, the methylation calling procedure was performed using the Python script bs_seeker2-call_methylation.py from Bsseeker2 using the default options. Additionally, methylated sites with fewer than ten reads mapped within the region were filtered out.

Differentially methylated loci (DMLs) and differentially methylated regions (DMRs) were identified using the R package DSS [26]. First, a simple average algorithm for smoothing was used to estimate mean methylation levels. The DMLs were identified by comparing the mean methylation levels in the NPR and C groups for each methylated site. The DMRs were identified based on regions harboring statistically significant methylated sites based on the following criteria: FDR-adjusted P value < 10–5 for methylated sites, minimum length of 50 bp, minimum number of 50 methylated sites, significant percentage of methylated sites being in the region (0.5), and a methylation difference greater than 0.1. Additionally, the DMRs mapped within regions located less than 50 bp from each other were merged into a single DMR. The DMRs were assigned to candidate genes and annotated for genomic context (promoter, intron, exon, and intergenic) with the R package genomation [27] using the gene annotation file from the ovine reference genome Oar_Ram_v2.0 (annotation release 104). For DMRs that were not mapped within a gene coordinate (intergenic regions), the closest gene was assigned to the DMR.

Gene Ontology and metabolic pathway enrichment analysis and quantitative trait locus annotation

The R package gprofiler2 was used for GO term and KEGG [28, 29] and Reactome pathway enrichment analysis. Enriched status was defined based on a false discovery rate (FDR) < 0.05. Only GO terms with more than 2 and fewer than 1000 assigned genes were considered for enrichment status. A redundancy reduction analysis was performed using the rutils package in R. The go_reduce() function was used to estimate semantic similarities between the GO terms via the Wang method, where a graph-based strategy for computing semantic similarity using the topology of the GO graph structure is applied. Subsequently, terms with a similarity threshold higher than 0.7 were grouped. The smallest adjusted P value was assigned for the different GO terms grouped under the same parental GO term, and duplicates were removed. The enrichment analysis was performed individually for DMRs with higher methylation means in each group (NPR and C). Additionally, the GALLO R package [30] was used to annotate the colocalization of the genes harboring DMRs with quantitative trait loci (QTL) using SheepQTLdb from Animal QTLdb [31]. An interval of 250 kb downstream and upstream (500 kb in total) from the start and end coordinates of each DMR was considered for QTL annotation. The gff file from SheepQTLdb (Oar_Ram_v2.0) was edited to remove QTLs with lengths greater than 1 Mb. This approach was chosen to avoid the annotation of QTLs covering large regions of chromosomes, usually identified by linkage methods and using low-density microsatellite marker maps.

Statistical analyses

The milk composition of the 28 ewes subjected to WGBS was recorded at 11 time points. The sampling points, represented as the mean (± standard deviation) days in milk were: 26.39 (± 8.02), 32.39 (± 8.02), 40.39 (± 8.02), 46.39 (± 8.02), 52.39 (± 8.02), 53.39 (± 8.02), 57.39 (± 8.02), 58.39 (± 8.02), 59.39 (± 8.02), 65.39 (± 8.02), 66.39 (± 8.02).

In all these points, the fat percentage (FP), protein percentage (PP), dry extract percentage (DE), SCC, and milk yield (MY) were assessed. Milk yield was measured by weighing the total milk produced by each animal during morning and evening milking. Milk FP, PP, and DE were determined by infrared spectrophotometry (ISO 9622:1999) using a MilkoScan FT6000 (Foss), combined with a fluoro-opto-electronic counter (Fossomatic 5000, Foss) for SCC (ISO 13366–2:2006). The average values of FP, PP, DE, and SCC were log-transformed and compared between the NPR and C groups using a generalized linear model, where the number of lambs born during the current lactation was included as a fixed effect. The MY values were maintained in the original scale and compared between NPR and C using the same model. The statistical analyses were performed using the statistical software R (R version 4.2.0) [32].

The Pearson correlation between the raw values of FP, PP, DE, SCC, and MY and the mean methylation level of each DMR was estimated using the cor.test function in R. The correlations were estimated for each milk trait (FP, PP, DE, SCC, and MY) individually for the samples of the NPR and C groups. Significant correlations were defined using the threshold of a P value < 0.05.

Results

Whole-genome bisulfite sequencing statistics

The mapping statistics for the reads obtained via WGBS are shown in Supplementary Table 2. An average mapping rate of 66.40 ± 3.32% was obtained, with values ranging from 59.70% to 69.10%. In eukaryotes, methylation can occur in three different contexts: CG, CHG, and CHH, where H is adenine (A), cytosine (C), or thymine (T). The mean percentages (± standard deviation) of methylated sites in CG, CHG and CHH contexts were 71.33 ± 1.15%, 1.34 ± 0.02%, and 1.42 ± 0.02%, respectively, in the NPR group. The means for the CG, CHG and CHH contexts for the control (C) group were 71.56 ± 0.97%, 1.34 ± 0.02%, and 1.41 ± 0.02%, respectively. Therefore, similar methylation patterns were observed in the NPR and C groups.

Identification of DMLs and DMRs between NPR and control ewes

In total, 32,247 DMLs were identified between the NPR and C groups. A circular Manhattan plot showing the distribution of the adjusted p values of the DMLs and a density plot showing the distribution of the significant DMLs across the genome is presented in Fig. 1. The most prominent association peaks were observed on chromosomes 1, 7, 11, 20, and 24. The calling of DMRs among these DMLs resulted in the identification of 1,154 DMRs (Supplementary Table 3). The distribution of these DMRs across chromosomes, their lengths and the corresponding mean methylation distribution are shown in Fig. 2A and B. The largest number of DMRs was observed on chromosome 1 (97 DMRs), and the smallest number of DMRs was identified on chromosome X (14 DMRs). The comparison of the lengths and mean methylation levels of the DMRs did not show substantial general differences between the NPR and C groups. Additionally, it is important to highlight that the mean methylation level was compared with the methylation level of hypermethylated DMRs in the NPR and C groups (Fig. 2C). Consequently, despite the general similarity between the distribution of the mean methylation level, all of these DMRs presented differential methylation patterns between the NPR and C groups. The percentage of DMRs annotated in each gene context (promoter, exon, intron, and intergenic) and the number of genes assigned exclusively or simultaneously to DMRs that were hypermethylated in the NPR or C group are shown in Fig. 2 D and E, respectively. The majority of DMRs were mapped to introns or intergenic regions, with promoters and exons corresponding to 8.64% and 15.86% of the DMR genomic context, respectively. Only 36 genes were simultaneously assigned to DMRs that were hypermethylated in the NPR and C groups. In total, 540 genes were exclusively assigned to DMRs that were hypermethylated in the NPR group, while 441 genes were exclusively assigned to DMRs that were hypermethylated in the C group.

Fig. 1
figure 1

Circular Manhattan plot and genomic density distribution of differentially methylated loci (DMLs) identified in the comparison between the nutritional protein restriction (NPR) and Control group. In the circular Manhattan plot, the associated values for each DML is in a -log10 scale. In the density plot, the darker the red shade on the bar plots, the highest the density of the DMLs within the 1 Mb windows comprising the DMLs. The legend of the density plot represents the number of DMLs associated with each color scale

Fig. 2
figure 2

Distribution of differentially methylated regions (DMRs) per chromosome (A), per length in base pairs, bp (B) and by mean methylation (C). The percentage of DMRs per each gene context (promoter in purple, exon in red, intron in green, and intergenic in blue) is shown in the pie plot (D). The number of genes shared between the DMRs hypermethylated for the nutritional protein restriction (NPR) or control group is shown in the Venn Diagram (E). For the violin plots and Venn Diagram, the mean methylation level for DMRs hypermethylated in the control group is displayed in green, and the mean methylation for those DMRs hypermethylated in the NPR group is indicated in red

Candidate genes and functions associated with DMRs identified between NPR and control ewes

The ten DMRs with the largest absolute AreaStat (the sum of the test statistics of all CpG sites within the DMR) are shown in Table 1. The regions harboring the most prominent peaks of DMLs (located on chromosomes 1, 11, 20, and 7) were scrutinized to identify the DMRs and the candidate genes in those regions. These regions were identified by a constant pattern of decreasing p values and false discovery rates (FDRs) < 1 × 10–100. Figure 3 shows the genomic context of the two regions harboring the largest number of DMRs on chromosomes 1 (5 DMRs in a 45.5 Kb interval from 112,851,283–112,896,784) and 20 (6 DMRs in a 1,155.25 Kb interval from 50,206,967–51,362,216). All DMRs in the abovementioned region of chromosome 1 were hypermethylated in the NPR group (Supplementary Fig. 2A). On the other hand, among the 6 DMRs called in the analyzed region of chromosome 20, four were hypermethylated, and 2 were hypomethylated in the NPR group (Supplementary Fig. 2B). The DMRs mapped on chromosome 1 are located in a region composed of several tRNAs and two LOCs (mRNA-basic proline-rich protein-like and mRNA-collagen alpha-1(I) chain-like). Additionally, a second region on chromosome 1 harbored one of the DMRs with the ten highest AreaStat values. The DMR mapped in this region (1:3,049,876–3050313) was also hypermethylated in the NPR group and mapped to exonic/intronic regions of PER2. From the DMRs mapped on chromosome 20, the two hypomethylated DMRs in the NPR group were assigned to the GMDS gene. Regarding the hypermethylated DMRs in the NPR group mapped on chromosome 20, two were assigned to the FOXC1 gene, one was assigned to the LOC114109593 gene, and one was assigned to the LOC121817372 gene. Additionally, two DMRs were called in the analyzed region of chromosome 7, one hypomethylated in the NPR group and one hypermethylated in the NPR group. Both DMRs were mapped to intronic regions of the TTC7B gene. On chromosome 11, only one DMR was called in the regions harboring prominent peaks of DMLs. This DMR was mapped to 11:42,240,935–42,241,421, which corresponds to the exonic region of the CAVIN1 gene. This DMR was hypomethylated in the NPR group (mean methylation = 0.518) compared to the C group (0.754).

Table 1 Differentially methylated regions (DMRs) with the ten highest absolute AreaStat (sum of the test statistics of all CpG sites within the DMR) identified in the comparison between the nutritional protein restriction (NPR) and control group
Fig. 3
figure 3

Bubble plots displaying the enrichment results for Gene Ontology (GO) terms (Biological Processes in black, Molecular Functions in blue, and Cellular Components in red) and metabolic pathways (in purple) obtained for the genes harboring hypermethylated DMRs in the control group (A) and nutritional protein restriction group (B). The area of the circles in the plot corresponds to the number of associated genes for that term, while the shade of red represents the adjusted P value (the darker the red shade, the smaller the P value is). The x-axis shows the richness factor, which corresponds to the ratio between the number of associated genes for a specific and the total number of genes assigned for this trait in the database

In total, 40 Gene Ontology (GO) terms and 2 KEGG pathways were enriched (FDR < 0.05) in genes assigned to the identified DMRs. One GO term was shared between the genes harboring DMRs that were hypermethylated or hypomethylated in the NPR group (ameboidal-type cell migration). The genes assigned to DMRs that were hypomethylated in the NPR group were associated with 20 enriched GO terms (16 biological processes and four cellular component terms). For the genes assigned to DMRs that were hypermethylated in the NPR group, 19 GO terms (15 biological processes, two molecular functions and two cellular components) and two KEGG pathways were identified as enriched. All enriched terms and pathways are shown in Fig. 3, and the complete enrichment results are available in Supplementary Table 4. In general, the enriched terms for the genes harboring DMRs that were hypomethylated in the NPR group (hypermethylated in the C group) were related to the regulation of organismal development, morphogenesis, and homeostasis. On the other hand, the DMRs that were hypermethylated in the NPR group were associated with more specialized enriched terms, such as phospholipid binding, fat cell differentiation, epithelial cell proliferation, circadian behavior, circadian regulation of gene expression, and negative regulation of cold-induced thermogenesis.

The milk-related quantitative trait loci (QTL) were the most frequent QTL class annotated within the coordinates (± 250 kb) of the DMRs that were hypermethylated in the NPR (33.13%), and C (29.87%) groups (Fig. 4). Enrichment analysis suggested that the trait assigned to each QTL class was enriched within the DMR coordinates evaluated (data not shown). For both NPR and C hypermethylated DMRs, the most frequent traits assigned to the annotated milk-related QTLs were milk fat yield {180d}, milk yield {180d}, milk protein yield {180d}, and cheese yield. A total of 68 genes were identified harboring DMRs mapped within the coordinates of milk-related QTLs (Supplementary Table 5).

Fig. 4
figure 4

Percentages of quantitative trait loci (QTL) category (upper part) annotated for the differentially methylated regions in a 250 Kb interval (downstream and upstream) hypermethylated in NPR (A) and control groups (B). The bar plots (lower part) show the percentage of each milk-related trait assigned to the milk QTL category

Correlations between milk production and differentially methylated regions

The mean values of FP, PP, DE, SCC, and MY for the 28 ewes evaluated in the current study are available in Supplementary Table 6. In comparisons between the NPR and C groups, one animal was excluded since it was the only ewe with a parity of four lambs and because this ewe showed substantially larger numbers of SCC across all 11 time points sampled. The comparison of the log-transformed values between the NPR and C groups indicated no effect of the dietary group on FP, PP, DE, SCC, or MY (Table 2).

Table 2 Mean (± standard deviation) values for fat, protein and dry extract percentages and number of somatic cells counts in the milk between the nutritional protein restriction (NPR) and control (C) groups

In total, 120, 128, 115, 151, and 118 DMRs were significantly correlated with FP, PP, DE, SCC and MY, respectively (Fig. 5A and Supplementary Table 7). The number of genes harboring DMRs correlated with different milk-related traits is shown in Fig. 5B. SCC was the trait with the largest number of unique genes harboring significantly correlated DMRs (108 genes). Five genes harbored DMRs that were correlated with all four milk traits: EPHA2, SHANK2, LOC114117507, LOC101121820, and LOC114110015. The enriched GO terms and/or metabolic pathways of the genes harboring DMRs that were significantly correlated with each trait individually are shown in Fig. 6 A-D, respectively. The complete list of enriched GO terms and metabolic pathways identified for the genes harboring DMRs correlated with FP, PP, DE, and SCC is available in Supplementary Table 8. There were no enriched GO terms or metabolic pathways among the genes harboring DMRs that were significantly correlated with FP based on the 5% FDR threshold. Therefore, in Fig. 6A, a 10% FDR threshold was adopted for identifying enriched terms. Additionally, just one GO term, “Ruffle membrane” (from the cellular component class), was identified as enriched for the genes positively correlated with MY even applying a FDR 10% threshold. The GO terms associated with the genes harboring DMRs that were significantly correlated with FP were related to phospholipid binding (among other processes). For the genes harboring DMRs correlated significanty with PP, important GO terms were observed, such as Cellular response to vascular endothelial growth factor stimulus, Response to muscle stretch, and Regulation of protein sumoylation. Regarding the GO terms enriched for the genes harboring DMR correlated with SCC, several terms associated with cellular proliferation and migration were observed (Regulation of endothelial cell migration, Epithelium migration, Epithelial cell proliferation, Regulation of cellular response to growth factor stimulus). Table 3 shows 48 DMRs that were significantly correlated with at least one of the milk traits (FP, PP, DE, and SCC) and were mapped within the genic regions (promoter, exon, and intron) of 44 genes associated with at least one of the enriched GO terms or metabolic pathways. Among these 48 DMRs, 18 significant correlations (11 negative and 7 positive) were observed for 17 DMRs mapped within 17 genes for FP. For PP, 18 significant correlations (5 negative and 13 positive) were observed for 18 DMRs mapped within 17 genes. The 14 significant correlations observed with DE (2 negatives and 12 positives) corresponded to 14 DMRs mapped within 13 genes. For SCC, 18 significant correlations (10 negatives and 8 positives) were observed, which were mapped within the genic regions of 17 genes harboring 17 DMRs associated with at least one of the enriched GO terms or metabolic pathways. Finally, for MY three significant correlations were observed between DMRs mapped in three different genes: FAM107A (0.543), PSD2 (-0.674), and TESC2 (-0.648).

Fig. 5
figure 5

Functional characterization of differentially methylated regions (DMR) significantly correlated with fat (FP), protein (PP), dry extract (DE) percentages, somatic cell counts (SCC), and milk yield (MY). A Number of DMRs positively (in red) and negatively (in green) correlated with each evaluated trait identified individually using the data comprising the nutritional protein restriction (NPR) and control group. B Venn diagram showing the number of genes harboring significantly correlated DMRs, shared between FP (in red), PP (in green), DE (in pink), and SCC (in purple). MY was not included in the Venn diagram because just one enriched GO term was identified for the genes harboring significantly correlated DMRs with MY

Fig. 6
figure 6

Bubble plots displaying the enrichment results for Gene Ontology (GO) terms (Biological Processes in black, Molecular Functions in blue, and Cellular Components in red) and metabolic pathways (in purple) obtained for the genes harboring significantly correlated DMRs with FP (A), PP (B), DE (C), and SCC (D). The area of the circles in the plot corresponds to the number of associated genes for that term, while the shade of red represents the adjusted p-value (the darker the red shade, the smallest the p-value). The x-axis shows the richness factor, which corresponds to the ratio between the number of associated genes for a specific and the total number of genes assigned for this trait in the database

Table 3 Differentially methylated regions (DMRs) identified in the comparison between the nutritional protein restriction (NPR) and control group mapped within gene coordinates (promoter, exon, and introns) identified in the comparison between NPR and control group and significantly correlated with fat (FP), protein (PP), dry extract (DE) percentages, somatic cells count (SCC), and milk yield (MY)

In general, DMRs that were hypermethylated in the NPR group were associated with enriched terms involved in biological processes that are functionally relevant to mammary gland production, such as phospholipid binding, fat cell differentiation, epithelial cell proliferation, the response to growth factors, and the regulation of circadian behavior. On the other hand, the hypermethylated DMRs in the C group (or hypomethylated DMRs in the NPR group) were associated with enriched GO terms related to organism homeostasis, the regulation of cell development, export from the cell, tissue development and synapsis-related processes. These processes may be related to the general homeostasis, development and activity of the mammary gland.

It is important to highlight that the number of significant correlations presented is larger than the number of DMRs because each DMR was tested for correlation with each of the considered milk production traits, and these correlations were also tested separately in the NPR and C groups.

Discussion

Epigenetic marks are relevant functional targets of nutritional stress conditions with important effects on the metabolic status of the organism [33,34,35]. Nutritional status is an important factor affecting milk production and quality in dairy sheep, in which fat-related characteristics tend to be more easily affected than milk protein levels [36]. Indeed, the supplementation of protein and amino acids only marginally affects milk protein levels [36]. Recently, our research group reported the absence of an effect of NPR on milk production traits such as SCC during the first lactation of Assaf ewes [16]. This analysis was performed in the same dataset of animals evaluated in the current study, where the absence of an effect of NPR in the prepubertal stage on FP, PP, DE, and SCC was also observed during the second lactation. Despite the absence of an effect on milk-related production traits, a relatively large number of DMRs (1154) were observed between the milk somatic cells of animals subjected to NPR or not. An important aspect to consider is that the substitution of soybean meal from the diet implies a modification in the qualitative composition of the NPR diet beyond the reduction of crude protein (42.3% in NPR animals). Therefore, observed effects on DNA methylation might not be solely attributed to protein restriction but also to (potential) differences in metabolizable energy intake and/or modified intake of other nutrients such as ADF, ADL or lipids.

On chromosome 1, two regions showed a pattern of increased significance for DMLs. Indeed, the DMRs mapped within these regions were among the top 10 DMRs with the highest absolute AreaStat values. The first region comprised 5 DMRs in a 45.5 Kb interval from 112,851,283–112,896,784, and it was mapped within a region with several annotated tRNAs. In lactating rats, perinatal protein restriction affects the free amino acid and fatty acid profile of the milk [37]. Pulina et al. (2006) [36] described divergent effects of diets with different protein contents on milk composition. The authors suggested that these contrasting results might be related to the milk production level achieved in those animals. The epigenetic regulation of genomic tRNA loci is described in the literature as an important mechanism regulating a plethora of biological processes, including metabolic regulation and age-related adaptations, and patterns of differential methylation in regions with clusters of several tRNAs have previously been described [38,39,40,41]. The other region on chromosome 1 harbored one DMR (1:3,049,876–3050313), which was mapped within exonic and intronic regions (depending on the transcript) of the PER2 gene. This DMR was more than twice as hypermethylated in the NPR group as in the C group. The PER2 gene is associated with the control of circadian rhythm in the central nervous system and peripheral organs [42]. An important function of PER2 is the regulation of PPARγ, a nuclear receptor that plays crucial roles in adipogenesis, the inflammatory response and insulin sensitivity [43,44,45]. In mice, PER2 deficiency results in drastic reductions in total triacylglycerol and nonesterified fatty acid levels in white adipocytes [46]. In sheep, the dietetic suppression of melatonin alongside constant exposure to light increases basal lipolysis and induces the overexpression of PER2 and PPARγ, among other adipogenic/thermogenic and circadian clock genes [47]. In addition, in cattle, the silencing of PER2 results in the suppression of lipid synthesis in the mammary gland through the regulation of SREBF1 and PPARγ [48]. The results obtained in the comparison between NPR and C animals for FP suggest potential differences between the groups that could not be observed here due to sample size limitations. The PER2 region is hypomethylated in the NPR animals in the current experiment, and it might be acting in the inhibition of its expression in the mammary gland of these animals, consequently affecting the FP in the milk samples. Among other functional candidate genes harboring DMRs in the most prominent regions analyzed in the current study, FOXC1 and CAVIN1 stand out. A DMR mapped in an intergenic region close to FOXC1 (20:50,694,985–50,695,939) was hypermethylated in the NPR group. In mice, the overexpression of FOXC1 results in the suppression of lobuloalveologenesis and lactation associated with higher percentages of estrogen receptor-, progesterone receptor-, or ki67-positive mammary epithelial cells during the lactation stage [49]. The epigenetic control of FOXC1 during mammary gland development was previously described in humans [50]. In the current study, one of the exons of the CAVIN1 gene was found to harbor a DMR (11:42,240,935–42,241,421) that was hypermethylated in the C group compared with the NPR group. This gene encodes Caveolae Associated Protein 1, which acts alongside caveolin-1 to create a unique lipid environment in caveolae [51]. Caveolae are domains localized on the cell surface in vertebrates that play important roles in cell migration and mechanoprotection [52, 53]. The activity and function of caveolin-1 are modulated by CAVIN1 in different processes, as observed for the oncogenic activity of caveolin-1 [54]. In addition, caveolin-1 is downregulated during lactation through the action of prolactin, and caveolin-1-deficient mice show accelerated mammary gland development and premature lactation [55, 56]. The same authors demonstrated that the recombinant overexpression of caveolin in HC11 cells could inhibit the prolactin-induced activation of β-casein promoter activity and synthesis [55]. Therefore, FOXC1 and CAVIN1 have the potential to emerge as functional candidate genes involved in the epigenetic control of mammary gland production in sheep.

The analysis of QTLs previously described in the regions harboring the DMRs in the NPR and C groups demonstrated that most annotated QTLs were related to milk traits. Among all milk-related QTLs, the regions harboring DMRs in the current study were more frequently associated with DE yield (protein and fat, mainly), MY and cheese yield. These results suggest a potential role of these DMRs in regulating biological processes associated with milk-related production traits.

To identify DMRs resulting from nutrient protein restriction with the potential to regulate milk-related production traits, we tested the correlations between the mean methylation levels within the 1154 DMRs and FP, PP, DE, SCC, and MY within the NPR and C groups individually. In total, DMRs mapped within 424 genes were significantly correlated with at least one of the milk traits evaluated. The phospholipid-binding molecular function was the most enriched GO term associated with the genes harboring DMRs correlated with FP. Phospholipids are crucial molecules that depend on coordinated flux and availability for the proper secretion of milk fat and other components [57]. The genes harboring DMRs correlated with PP resulted in the largest number of enriched GO terms. Among these terms, the large number of terms associated with the development of different structures, such as the differentiation of mesenchymal cells and epithelium migration, can be highlighted. The interactions between mesenchymal and epithelial cells are crucial for the proper development of the mammary gland [58]. The Rap1 metabolic signaling pathway was the most enriched for those genes harboring DMRs correlated with DE. Recently, Rap1 signaling was identified as being enriched in coexpressed gene networks from bovine mammary epithelial cells contrasted by high- and low-fat rates [59]. Additionally, the Rap1 signaling pathway is an important component of the proliferation and differentiation of mammary epithelial cells with the potential to increase milk production [60,61,62]. The number of enriched terms linked to endothelial and epithelial cell proliferation and migration associated with the genes harboring DMRs correlated with SCC stands out. The epithelial cells that are shed into milk during lactation have cellular characteristics of terminally differentiated luminal cells, and the analysis of immunohistochemical markers for proliferation indicated an increase in epithelial cell proliferation from early lactation to late lactation in cows [63]. This finding is corroborated by the intense renewal of epithelial cells during lactation, where most mammary cells present in the mammary gland at the end of lactation are formed after calving in dairy cows [64]. The involvement of the genes harboring DMRs correlated with milk production traits in biological processes crucial for mammary gland development and production reinforces the possibility that these epigenetic markers act in a compensatory mechanism related to the protein restriction to which the animals were subjected. Only five genes harbored DMRs correlated with all four milk traits, among which three were uncharacterized loci: LOC114117507, LOC101121820, and LOC114110015, while the other two genes were EPHA2 and SHANK2. SHANK2 encodes one of the major scaffold proteins of excitatory synapses [65]. The establishment of epithelial cell polarity is controlled by the formation of tight junctions with the participation of the Rap1 signaling pathway through the binding of SHANK2 to atypical protein kinase C [66]. The DMR mapped in the intronic region of SHANK2 (21:44,415,614–44,415,779) was hypermethylated under NPR and was positively correlated with FP, PP, and DE, while it was negatively correlated with SCC. EPHA2 has been demonstrated to act as an important component of mammary gland development, with roles in epithelial proliferation and branching morphogenesis, a process that is more active during puberty [67]. A DMR mapped to an intergenic region close to EPHA2 (2:249,637,801–249,638,050) was hypermethylated in the NPR group compared with the C group and showed negative correlations with FP, PP, and DE. In contrast, a positive correlation was observed with SCC. The highest absolute correlations were observed between the DMR (4:121,076,547–121,076,925) mapped to the intronic region of the ESYT2 gene and DE and FP. ESYT2 encodes Extended Synaptotagmin 2, a member of the Synaptotagmin complex, which acts together with SNARE (Soluble N-Ethylmaleimide-Sensitive Factor Attachment Protein Receptor) proteins as a mediator of the specific fusion of transport vesicles and exocytosis with potential functions in mammary epithelial cells [68]. Additionally, ESYT2 controls the dynamics of Ca2+ in different types of cells [69], and it is suggested to play a direct role in lipid transport [70]. The DMR mapped in the intronic region of ESYT2 (4:121,076,547–121,076,925) was hypermethylated in the C group compared with the NPR group, and it was positively correlated with DE and FP.

Among the other genes harboring DMRs that were significantly correlated with milk traits, the functional impact of GATA3, HMGB1, CTBP2, and TOM1L2 can be highlighted. GATA3 encodes a transcription factor expressed in luminal breast epithelial cells that regulates cell proliferation in this tissue [71]. In the current study, a DMR in GATA3 (13:12,393,887–12,393,956) was hypermethylated in the C group and negatively associated with SCC according to the values from the animals of the C group. In mice, changes in the expression of HMGB1, encoding the high mobility group box 1 protein, are observed during mammary gland development, with lower values occurring during lactation and involution [72]. Here, a hypermethylated DMR in the C group (10:30,611,368–30611452), mapped within HMGB1, was positively correlated with SCC. CTBP2 genes have previously been linked with transcriptional corepressor activity in the liver, acting as metabolic sensors responsible for regulating glucose and lipid homeostasis [73]. Three different DMRs in CTBP2 were positively correlated with DE (22:44,176,758–44,176,942 and 22:44,085,601–44,085,731), FP (22:44,085,601–44,085,731) and PP (22:44,114,597–44,114,726). All of these positive correlations were observed in relation to the values obtained for these traits in the C group samples. Additionally, one DMR in CTBP2 (22:44,193,718–44,193,809) was negatively correlated with the PP values of the samples in the NPR group. TOM1L2 encodes a protein involved in membrane trafficking and endocytosis [74, 75]. In goats, hypomethylated DMR on TOM1L2 was previously identified by comparing mammary gland samples from dry and lactation periods [76]. Here, a DMR in the intronic region of TOM1L2 (11:34,328,017–34,328,106), which was hypermethylated in the C group, was positively correlated with DE and FP. We also highlight that SREBF1 is a major regulator of lipid metabolism in the mammary gland, and a putative regulatory effect of this DMR on SREBF1 cannot be disregarded [77,78,79]. Despite the absence of functionally relevant enriched terms for the DMRs correlated with MY, it is interesting to highlight an intronic DMR (9:21,489,061.21489139) mapped in the Thyroglobulin (TG) gene which was negatively correlated with FP (-0.538) and positively correlated with MY (0.622). Polymorphims in the TG were previously associated with 305-day milk fat percent and otal lactation fat percentage in dairy cows and buffalo [80, 81].

The results obtained here pinpoint putative functional candidate genes for milk production traits in sheep based on differential methylation patterns. Future studies can leverage the results obtained here to better evaluate the regulatory elements within the DMRs described in the current study. The analysis of active promoters and enhancers for these functional candidate genes has the potential to better characterize the regulatory mechanisms enrolled with the expression of those genes and the phenotypic variance observed among individuals [82]. Additionally, analysis comprising the methylation level of the candidate DMRs reported here for each cellular type available in milk samples might help to understand the contribution of each cell type to production traits. It is important to mention that the methylation status can be a dynamic process and change across time and development. For example, in sheep, the analysis of differential methylation profiles between fetal and adult muscular tissue was useful in providing new insights regarding the development of these tissues and the identification of potential functional candidate genes that might be associated with meat quality and production-related traits [83]. Therefore, a longitudinal analysis of DMRs in the milk somatic cells across lactations would help to identify time-specific methylation marks and candidate genes associated with milk components and productions in sheep. Despite the absence of significant differences between the NPR and C groups for the milk-related traits evaluated here, it is important to mention that these animals showed a significant difference in the body weight at the end of the nutritional protein restriction trial as shown in Pelayo et al. (2023) [84]. The data evaluated here is composed by a limited set of phenotypes at the second lactation of these animals. Therefore, a longitudinal study evaluating the association of the DMRs described here with the same phenotypes (and additional milk-related traits) is crucial to better understand the functionality of these methylation marks.

The mammary gland is a complex organ that provides a combination of immune and epithelial cells. The milk somatic cells are mainly cells from the immune system, such as lymphocytes, macrophages, and polynuclear cells. The reasons for using milk somatic cells in the current studies can be classified into practical, evidence-based, and animal welfare. The animal welfare aspect is related to the global demands for animal research, which are based, among other aspects, on the 3Rs (Replacement, Reduction, and Refinement) principle. These principles are considered a systematic approach to animal experimentation that puts the well-being of the animals front and center. Based on the 3Rs recommendations, an invasive approach (mammary-gland biopsy) should be replaced when an efficient proxy is available (Milk somatic cells analysis). The evaluation of mammary epithelial cells requires the application of invasive methodologies, such as mammary biopsies. From the practical point of view, mammary biopsies are not viable in commercial flocks, which is the case of the animals used in the current study, due to potential mammary tissue damage and disruptions in the lactation process. One of the main goals of the current study is to identify potential biomarkers that could be evaluated in commercial herds. Therefore, the use of milk somatic cells is a viable alternative for the evaluation of such biomarkers. Indeed, from an evidence-based point of view, despite the majority of the milk somatic cells being immune cells, in goats and sheep, several studies described an association between milk somatic cells and different milk production traits, such as milk yield, protein, mineral, and fat contents, and cheese making [85]. The use of WGBS for the investigation of genetic mechanisms associated with milk production is a relatively new approach in all livestock species, especially in sheep. Therefore, there are not studies showing the actual correlation between epigenetic markers in milk somatic cells and mammary gland tissues. However, together with the correlations between milk somatic cells and milk production traits described above, some additional information reinforces the viability of using milk somatic cells for the functional evaluation of the mammary gland. For example, studies in other species showed that using the transcriptome from milk somatic cells is a good representation of the mammary gland transcriptome obtained from biopsies [86,87,88,89]. Furthermore, in a study conducted in our research group [90], we proved that transcriptome analysis of somatic cells in milk from healthy animals is an excellent proxy to analyze specific functional changes in mammary gland epithelial cells (changes in the expression profile of genes related to the de novo synthesis of milk fat and protein) in response to specific feeding strategies or challenges such as milk fat depression. Additionally, the evaluation of specific biomarkers for mammary gland activity, such as the stearoyl‐CoA desaturase (SCD1) expression, suggested milk somatic cells of lactating cows as an indicator of SCD1 activity within the mammary gland [90]. This conclusion was reached due to the high correlation between the expression values of SCD1 between the tissues and also because the SCD1 expression in milk somatic cells was significantly related to Δ9-desaturase indices in milk, which are commonly used as an indicator of SCD1 activity within the mammary gland [87]. Despite the absence of a study evaluating the different methylation levels between the different cell types present in the milk somatic cells, some interesting comparisons can be made from results obtained from human blood samples. Similarly to the milk somatic cells, whole blood samples are also composed by different cellular types (in different proportions). Talens et al. (2010) [91] analyzed the variations of DNA methylation in different human tissues, with an exciting focus on the correlations between the methylation profile of different whole blood cellular types. The results obtained suggested that the majority of variation observed in the methylation pattern was not explained by the cellular heterogeneity, and when some variation in DNA methylation that could be explained by variation in cellular heterogeneity was observed, this variation was generally small, and associations were of borderline significance. Therefore, based on the abovementioned evidences, we consider the use of milk somatic cells an interesting alternative for the epigenetic evaluation of the sheep mammary gland.

Conclusion

The results presented here suggest a relevant effect of DMRs on the expression of genes associated with milk production and lipid metabolism in sheep. The DMRs were identified using a strict statistical threshold, and the experimental groups were distinguished from each other by protein restriction in the diet during the prepubertal stage. Although it is impossible to disregard effects on other milk characteristics, such as fatty acid and free amino acid profiles, there were no longer-term effects (on the second lactation) of protein restriction on FP, PP, DE, and SCC. Consequently, the abovementioned results might suggest a potential impact of the DMRs mapped within these candidate genes responsible for regulating key components of mammary gland development and production. It is important to highlight that the absence of a significant correlation of some of the DMRs with the evaluated milk traits did not exclude the possibility of a functional impact of these epigenetic markers in the mammary gland. In light of the above, the results obtained from the current study help to better understand the epigenetic mechanisms associated with milk production in the mammary gland of dairy sheep. The functionality of these epigenetic markers could be validated in further studies applying techniques such as ATAC-seq and the identification of expression QTLs (eQTLs) associated with the DMRs identified herein and other milk-related production traits.

Availability of data and materials

All sequence data obtained in the current study were uploaded to the European Nucleotide Archive under the accession number PRJEB56589 (https://www.ebi.ac.uk/ena/browser/view/PRJEB56589).

Abbreviations

C:

Control group

DE:

Dry extract percentage

DML:

Differentially methylated loci

DMR:

Differentially methylated regions

FDR:

False-discovery rate

FP:

Fat percentage

NPR:

Nutritional protein restriction group

PP:

Protein percentage

QTL:

Quantitative trait loci

SCC:

Somatic cell counts

WGBS:

Whole genome bisulfite sequencing

References

  1. United Nations. Revision of World Population Prospects. United Nations. 2019;

  2. Maja MM, Ayano SF. The Impact of Population Growth on Natural Resources and Farmers’ Capacity to Adapt to Climate Change in Low-Income Countries. Earth Systems and Environment. 2021.

  3. United Nations Food and Agriculture Organization. The State of Food and Agriculture 2016 (SOFA): Climate change, agriculture and food security. Livestock in the Balance. 2016.

  4. Connor EE. Invited review: Improving feed efficiency in dairy production: Challenges and possibilities. Animal. 2015;9:395–408.

    CAS  PubMed  Google Scholar 

  5. Ho CKM, Malcolm B, Doyle PT. Potential impacts of negative associative effects between concentrate supplements, pasture and conserved forage for milk production and dairy farm profit. Anim Prod Sci. 2013;53.

  6. Taelman SE, de Meester S, van Dijk W, da Silva V, Dewulf J. Environmental sustainability analysis of a protein-rich livestock feed ingredient in the Netherlands: Microalgae production versus soybean import. Resour Conserv Recycl. 2015;101.

  7. Nudda A, Atzori AS, Correddu F, Battacone G, Lunesu MF, Cannas A, et al. Effects of nutrition on main components of sheep milk. Small Ruminant Research. 2020;184.

  8. Sinha YN, Tucker HA. Mammary Development and Pituitary Prolactin Level of Heifers from Birth through Puberty and during the Estrous Cycle. J Dairy Sci. 1969;52:507–12.

    CAS  PubMed  Google Scholar 

  9. Anderson RR. Mammary Gland Growth in Sheep. J Anim Sci [Internet]. 1975 [cited 2022 Dec 25];41:118–23. Available from: https://academic.oup.com/jas/article/41/1/118/4668118

  10. Hassan A, Hamouda IA. Growth and Biochemical Changes in Mammary Glands of Ewes from 1 to 18 Months of Age. J Dairy Sci. 1985;68:1647–51.

    CAS  PubMed  Google Scholar 

  11. Hughes K. Comparative mammary gland postnatal development and tumourigenesis in the sheep, cow, cat and rabbit: Exploring the menagerie. Semin Cell Dev Biol. 2021;114:186–95.

    PubMed  Google Scholar 

  12. Smith GH. Binuclear Cells in the Lactating Mammary Gland: New Insights on an Old Concept? J Mammary Gland Biol Neoplasia. 2016.

  13. Villeneuve L, Cinq-Mars D, Lacasse P. Effects of restricted feeding of prepubertal ewe lambs on growth performance and mammary gland development. Animal. 2010;4:944–50.

    CAS  PubMed  Google Scholar 

  14. Silva AL, Detmann E, Dijkstra J, Pedroso AM, Silva LHP, Machado AF, et al. Effects of rumen-undegradable protein on intake, performance, and mammary gland development in prepubertal and pubertal dairy heifers. J Dairy Sci. 2018;101:5991–6001.

    CAS  PubMed  Google Scholar 

  15. Pirlo G, Capelletti M, Marchetto G. Effects of Energy and Protein Allowances in the Diets of Prepubertal Heifers on Growth and Milk Production. J Dairy Sci. 1997;80:730–9.

    CAS  PubMed  Google Scholar 

  16. Pelayo R., Marina H., Suarez-Vega A., Esteban-Blanco C., Foucras G., Arranz J. J., Gutierrez-Gil, B. Influence of a nutritional restriction in dairy ewe lambs on the response to a later inflammatory intramammary challenge. Proceeding of 12th World Congress on Genetics Applied to Livestock Production (WCGALP) Technical and species orientated innovations in animal breeding, and contribution of genetics to solving societal challenges. 2020; https://doi.org/10.3920/978-90-8686-940-4.

  17. Lillycrop KA, Hoile SP, Grenfell L, Burdge GC. DNA methylation, ageing and the influence of early life nutrition. Proc Nutr Soc. 2014;73(3):413–21.

    CAS  PubMed  Google Scholar 

  18. Kim K chol, Friso S, Choi SW. DNA methylation, an epigenetic mechanism connecting folate to healthy embryonic development and aging. Journal of Nutritional Biochemistry. 2009

  19. Ford D, Ions LJ, Alatawi F, Wakeling LA. The potential role of epigenetic responses to diet in ageing. Proceedings of the Nutrition Society. 2011.

  20. Fonseca PAS, Alonso-Garc\’\ia M, Pelayo R, Marina H, Esteban-Blanco C, Mateo J, et al. Integrated analyses of the methylome and transcriptome to unravel sex differences in the perirenal fat from suckling lambs. Front Genet. :3141.

  21. Andrews S. FASTQC A Quality Control tool for High Throughput Sequence Data. Babraham Institute. 2015;

  22. Krueger F. Trim Galore!: A wrapper tool around Cutadapt and FastQC to consistently apply quality and adapter trimming to FastQ files. Babraham Institute. 2015;

  23. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9:357–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Guo W, Fiziev P, Yan W, Cokus S, Sun X, Zhang MQ, et al. BS-Seeker2: A versatile aligning pipeline for bisulfite sequencing data. BMC Genomics. 2013;14:774.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25(16):2078–9.

    PubMed  PubMed Central  Google Scholar 

  26. Feng H, Conneely KN, Wu H. A Bayesian hierarchical model to detect differentially methylated loci from single nucleotide resolution sequencing data. Nucleic Acids Res. 2014;42:e69.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Akalin A, Franke V, Vlahoviček K, Mason CE, Schübeler D. Genomation: A toolkit to summarize, annotate and visualize genomic intervals. Bioinformatics. 2015;31(7):1127–9.

    CAS  PubMed  Google Scholar 

  28. Kanehisa M, Goto S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 2000 [cited 2023 Mar 1];28:27. Available from: /pmc/articles/PMC102409/

  29. Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 2015 [cited 2023 Mar 2];44:457–62. Available from: http://dailymed.nlm.nih.gov/

  30. Fonseca PAS, Suárez-Vega A, Marras G, Cánovas Á. GALLO: An R package for genomic annotation and integration of multiple data sources in livestock for positional candidate loci. Gigascience. 2020;

  31. Hu ZL, Park CA, Reecy JM. Building a livestock genetic and genomic information knowledgebase through integrative developments of Animal QTLdb and CorrDB. Nucleic Acids Res. 2019;47(D1):D701–10.

    CAS  PubMed  Google Scholar 

  32. R Core Team. R core team (2021). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. URL http://www.R-project.org.2021.

  33. Gomez-Verjan JC, Barrera-Vázquez OS, García-Velázquez L, Samper-Ternent R, Arroyo P. Epigenetic variations due to nutritional status in early-life and its later impact on aging and disease. Clin Genet. 2020;98:313–21.

    CAS  PubMed  Google Scholar 

  34. Anderson OS, Sant KE, Dolinoy DC. Nutrition and epigenetics: An interplay of dietary methyl donors, one-carbon metabolism and DNA methylation. Jo Nutr Biochem. 2012;23(8):853–9.

    CAS  Google Scholar 

  35. Etchegaray JP, Mostoslavsky R. Interplay between Metabolism and Epigenetics: A Nuclear Adaptation to Environmental Changes. Mol Cell. 2016;62(5):695–711.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Pulina G, Nudda A, Battacone G, Cannas A. Effects of nutrition on the contents of fat, protein, somatic cells, aromatic compounds, and undesirable substances in sheep milk. Anim Feed Sci Technol. 2006;131.

  37. Agnoux AM, Antignac JP, Boquien CY, David A, Desnots E, Ferchaud-Roucher V, et al. Perinatal protein restriction affects milk free amino acid and fatty acid profile in lactating rats: Potential role on pup growth and metabolic status. J Nutr Biochem. 2015;26:784–95.

    Google Scholar 

  38. Ebersole T, Kim JH, Samoshkin A, Kouprina N, Pavlicek A, White RJ, et al. tRNA genes protect a reporter gene from epigenetic silencing in mouse cells. Cell Cycle. 2011;10:2779–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Acton RJ, Yuan W, Gao F, Xia Y, Bourne E, Wozniak E, et al. The genomic loci of specific human tRNA genes exhibit ageing-related DNA hypermethylation. Nat Commun. 2021;12:2655.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Hummel G, Berr A, Graindorge S, Cognat V, Ubrig E, Pflieger D, et al. Epigenetic silencing of clustered tRNA genes in Arabidopsis. Nucleic Acids Res. 2020;48:10297–1031.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Hummel G, Warren J, Drouard L. The multi-faceted regulation of nuclear tRNA gene transcription. IUBMB Life. 2019;71(8):1099–108.

    CAS  PubMed  Google Scholar 

  42. Nishide SY, Hashimoto K, Nishio T, Honma KI, Honma S. Organ-specific development characterizes circadian clock gene Per2 expression in rats. Am J Physiol Regul Integr Comp Physiol. 2014;306:R67-74.

    CAS  PubMed  Google Scholar 

  43. Wafer R, Tandon P, Minchin JEN. The role of peroxisome proliferator-activated receptor gamma (PPARG) in adipogenesis: Applying knowledge from the fish aquaculture industry to biomedical research. Front Endocrinol (Lausanne). 2017.

  44. Picard F AJ. PPAR(gamma) and glucose homeostasis. Annu Rev Nutr. 2002;22.

  45. Janani C, Ranjitha Kumari BD. PPAR gamma gene - A review. Diabetes and Metabolic Syndrome: Clinical Research and Reviews. 2015.

  46. Grimaldi B, Bellet MM, Katada S, Astarita G, Hirayama J, Amin RH, et al. PER2 controls lipid metabolism by direct regulation of PPARγ. Cell Metab. 2010;12:509–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Seron-Ferre M, Reynolds H, Mendez N, Mondaca M, Valenzuela FJ, R E, et al. Impact of maternal melatonin suppression on amount and functionality of brown adipose tissue (BAT) in the newborn sheep. Front Endocrinol (Lausanne). 2014;5.

  48. Jing Y, Chen Y, Wang S, Ouyang J, Hu L, Yang Q, et al. Circadian gene per2 silencing downregulates pparg and srebf1 and suppresses lipid synthesis in bovine mammary epithelial cells. Biology (Basel). 2021;10:1226.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Gao B, Qu Y, Han B, Nagaoka Y, Katsumata M, Deng N, et al. Inhibition of lobuloalveolar development by FOXC1 overexpression in the mouse mammary gland. Sci Rep. 2017;7:14017.

    PubMed  PubMed Central  Google Scholar 

  50. Bloushtain-Qimron N, Yao J, Snyder EL, Shipitsin M, Campbell LL, Mani SA, et al. Cell type-specific DNA methylation patterns in the human breast. Proc Natl Acad Sci U S A. 2008;105:14076–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhou Y, Ariotti N, Rae J, Liang H, Tillu V, Tee S, et al. Caveolin-1 and cavin1 act synergistically to generate a unique lipid environment in caveolae. J Cell Biol. 2021;220:e202005138.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Nassoy P, Lamaze C. Stressing caveolae new role in cell mechanics. Trends Cell Biol. 2012;22(7):381–9.

    PubMed  Google Scholar 

  53. Parton RG. Caveolae: Structure, Function, and Relationship to Disease. Annu Rev Cell Dev Biol. 2018;34:111–36.

    CAS  PubMed  Google Scholar 

  54. Liu L, Xu HX, Wang WQ, Wu CT, Chen T, Qin Y, et al. Cavin-1 is essential for the tumor-promoting effect of caveolin-1 and enhances its prognostic potency in pancreatic cancer. Oncogene. 2014;33:2728–36.

    CAS  PubMed  Google Scholar 

  55. Park DS, Lee H, Riedel C, Hulit J, Scherer PE, Pestell RG, et al. Prolactin Negatively Regulates Caveolin-1 Gene Expression in the Mammary Gland during Lactation, via a Ras-dependent Mechanism. J Biol Chem. 2001;276:48389–97.

    CAS  PubMed  Google Scholar 

  56. Park DS, Lee H, Frank PG, Razani B, Nguyen Av, Parlow AF, et al. Caveolin-1-deficient mice show accelerated mammary gland development during pregnancy, premature lactation, and hyperactivation of the Jak-2/STAT5a signaling cascade. Mol Biol Cell. 2002;13:3416–30.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Smoczyński M. Role of Phospholipid Flux during Milk Secretion in the Mammary Gland. J Mammary Gland Biol Neoplasia. 2017;22(2):117–29.

    PubMed  PubMed Central  Google Scholar 

  58. Cunha GR, Hom YK. Role of mesenchymal-epithelial interactions in mammary gland development. J Mammary Gland Biol Neoplasia. 1996;1(1):21–35.

    CAS  PubMed  Google Scholar 

  59. Mu T, Hu H, Ma Y, Wen H, Yang C, Feng X, et al. Identifying key genes in milk fat metabolism by weighted gene co-expression network analysis. Sci Rep. 2022;12:1–13.

    Google Scholar 

  60. Itoh M, Nelson CM, Myers CA, Bissell MJ. Rap1 integrates tissue polarity, lumen formation, and tumorigenic potential in human breast epithelial cells. Cancer Res. 2007;67(10):4759–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Fata JE, Mori H, Ewald AJ, Zhang H, Yao E, Werb Z, et al. The MAPKERK-1,2 pathway integrates distinct and antagonistic signals from TGFα and FGF7 in morphogenesis of mouse mammary epithelium. Dev Biol. 2007;306:193–207.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Farhadian M, Rafat SA, Panahi B, Mayack C. Weighted gene co-expression network analysis identifies modules and functionally enriched pathways in the lactation process. Sci Rep. 2021;11:2367.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Baratta M, Volpe MG, Nucera D, Gabai G, Guzzo N, Faustini M, et al. Differential expression of living mammary epithelial cell subpopulations in milk during lactation in dairy cows. J Dairy Sci. 2015;98:6897–904.

    CAS  PubMed  Google Scholar 

  64. Capuco AV, Wood DL, Baldwin R, Mcleod K, Paape MJ. Mammary cell number, proliferation, and apoptosis during a bovine lactation: Relation to milk production and effect of bST. J Dairy Sci. 2001;84:2177–87.

    CAS  PubMed  Google Scholar 

  65. Sala C, Vicidomini C, Bigi I, Mossa A, Verpelli C. Shank synaptic scaffold proteins: Keys to understanding the pathogenesis of autism and other synaptic disorders. J Neurochem. 2015;135(5):849–58.

    CAS  PubMed  Google Scholar 

  66. Sasaki K, Kojitani N, Hirose H, Yoshihama Y, Suzuki H, Shimada M, et al. Shank2 Binds to aPKC and Controls Tight Junction Formation with Rap1 Signaling during Establishment of Epithelial Cell Polarity. Cell Rep. 2020;31:107407.

    CAS  PubMed  Google Scholar 

  67. Vaught D, Chen J, Brantley-Sieders DM. Regulation of mammary gland branching morphogenesis by EphA2 receptor tyrosine kinase. Mol Biol Cell. 2009;20:2572–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Truchet S, Ollivier-Bousquet M. Mammary gland secretion: Hormonal coordination of endocytosis and exocytosis. Animal. 2009;3:1733–42.

    CAS  PubMed  Google Scholar 

  69. Woo JS, Sun Z, Srikanth S, Gwack Y. The short isoform of extended synaptotagmin-2 controls Ca2+ dynamics in T cells via interaction with STIM1. Sci Rep. 2020;10:14433.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Schauder CM, Wu X, Saheki Y, Narayanaswamy P, Torta F, Wenk MR, et al. Structure of a lipid-bound extended synaptotagmin indicates a role in lipid transfer. Nature. 2014;510:552–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Emmanuel N, Lofgren KA, Peterson EA, Meier DR, Jung EH, Kenny PA. Mutant GATA3 actively promotes the growth of normal and malignant mammary cells. Anticancer Res. 2018;38(8):4435–41.

    PubMed  PubMed Central  Google Scholar 

  72. Brezniceanu ML, Völp K, Bösser S, Solbach C, Lichter P, Joos S, et al. HMGB1 inhibits cell death in yeast and mammalian cells and is abundantly expressed in human breast carcinoma. FASEB J. 2003;17:1295–7.

    CAS  PubMed  Google Scholar 

  73. Sekiya M, Kainoh K, Sugasawa T, Yoshino R, Hirokawa T, Tokiwa H, et al. The transcriptional corepressor CtBP2 serves as a metabolite sensor orchestrating hepatic glucose and lipid homeostasis. Nat Commun. 2021;12:6315.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Lohi O, Lehto VP. VHS domain marks a group of proteins involved in endocytosis and vesicular trafficking. FEBS Lett. 1998;440(3):255–7.

    CAS  PubMed  Google Scholar 

  75. Wang T, Liu NS, Seet LF, Hong W. The emerging role of VHS domain-containing Tom1, Tom1L1 and Tom1L2 in membrane trafficking. Traffic. 2010;11(9):1119–28.

    CAS  PubMed  Google Scholar 

  76. Zhang X, Zhang S, Ma L, Jiang E, Xu H, Chen R, et al. Reduced representation bisulfite sequencing (RRBS) of dairy goat mammary glands reveals DNA methylation profiles of integrated genome-wide and critical milk-related genes. Oncotarget. 2017;8(70):115326–44.

    PubMed  PubMed Central  Google Scholar 

  77. Rincon G, Islas-Trejo A, Castillo AR, Bauman DE, German BJ, Medrano JF. Polymorphisms in genes in the SREBP1 signalling pathway and SCD are associated with milk fatty acid composition in Holstein cattle. J Dairy Res. 2012;79:66–75.

    CAS  PubMed  Google Scholar 

  78. Han LQ, Gao TY, Yang GY, Loor JJ. Overexpression of SREBF chaperone (SCAP) enhances nuclear SREBP1 translocation to upregulate fatty acid synthase (FASN) gene expression in bovine mammary epithelial cells. J Dairy Sci. 2018;101:6523–31.

    CAS  PubMed  Google Scholar 

  79. Wickramasinghe S, Rincon G, Islas-Trejo A, Medrano JF. Transcriptional profiling of bovine milk using RNA sequencing. BMC Genomics. 2012;13:45.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Anton I, Kovács K, Holló G, Farkas V, Szabó F, Egerszegi I, et al. Effect of DGAT1, leptin and TG gene polymorphisms on some milk production traits in different dairy cattle breeds in Hungary. Arch Anim Breed. 2012;55.

  81. Dubey PK, Goyal S, Mishra SK, Yadav AK, Kathiravan P, Arora R, et al. Association analysis of polymorphism in thyroglobulin gene promoter with milk production traits in riverine buffalo (Bubalus bubalis). Meta Gene. 2015;5:157–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Davenport KM, Massa AT, Bhattarai S, McKay SD, Mousel MR, Herndon MK, et al. Characterizing Genetic Regulatory Elements in Ovine Tissues. Front Genet. 2021;12:628849.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Fan Y, Liang Y, Deng K, Zhang Z, Zhang G, Zhang Y, et al. Analysis of DNA methylation profiles during sheep skeletal muscle development using whole-genome bisulfite sequencing. BMC Genomics. 2020;21:327.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Pelayo R, Marina H, Suárez-Vega A, Hervás G, Esteban-Blanco C, Gausseres B, et al. Influence of a temporary restriction of dietary protein in prepubertal ewe lambs on first lactation milk traits and response to a mammary gland inflammatory challenge. Res Vet Sci. 2023;159:57–65.

    CAS  PubMed  Google Scholar 

  85. Raynal-Ljutovac K, Pirisi A, de Crémoux R, Gonzalo C. Somatic cells of goat and sheep milk: Analytical, sanitary, productive and technological aspects. Small Ruminant Research. 2007;68.

  86. Cánovas A, Rincón G, Bevilacqua C, Islas-Trejo A, Brenaut P, Hovey RC, et al. Comparison of five different RNA sources to examine the lactating bovine mammary gland transcriptome using RNA-Sequencing. Sci Rep. 2014;4:5297.

    PubMed  PubMed Central  Google Scholar 

  87. Jacobs AAA, Dijkstra J, Hendriks WH, van Baal J, van Vuuren AM. Comparison between stearoyl-CoA desaturase expression in milk somatic cells and in mammary tissue of lactating dairy cows. J Anim Physiol Anim Nutr (Berl). 2013;97:353–62.

    CAS  PubMed  Google Scholar 

  88. Boutinaud M, Rulquin H, Keisler DH, Djiane J, Jammes H. Use of somatic cells from goat milk for dynamic studies of gene expression in the mammary gland. J Anim Sci. 2002;80:1258–69.

    CAS  PubMed  Google Scholar 

  89. Murrieta CM, Hess BW, Scholljegerdes EJ, Engle TE, Hossner KL, Moss GE, et al. Evaluation of milk somatic cells as a source of mRNA for study of lipogenesis in the mammary gland of lactating beef cows supplemented with dietary high-linoleate safflower seeds. J Anim Sci. 2006;84:2399–405.

    CAS  PubMed  Google Scholar 

  90. Toral PG, Hervás G, Suárez-Vega A, Arranz JJ, Frutos P. Isolation of RNA from milk somatic cells as an alternative to biopsies of mammary tissue for nutrigenomic studies in dairy ewes. J Dairy Sci. 2016;99:8461–71.

    CAS  PubMed  Google Scholar 

  91. Talens RP, Boomsma DI, Tobi EW, Kremer D, Jukema JW, Willemsen G, et al. Variation, patterns, and temporal stability of DNA methylation: considerations for epigenetic epidemiology. The FASEB Journal. 2010;24.

Download references

Acknowledgements

Not applicable.

Funding

This work has received funding from the European Union’s Horizon 2020 research and innovation programme under Grant Agreement No 772787 (SMARTER). PASF is the beneficiary of a Maria Zambrano Grant of the University of Leon funded by the Ministry of Universities (Madrid, Spain) and financed by the European Union-Next Generation EU.

Author information

Authors and Affiliations

Authors

Contributions

PASF: Formal data analysis, Writing – original draft. ASV: Conceptualization, Methodology, Writing – review & editing. CEB: laboratory experiments and analysis, RP: laboratory experiments and analysis, HM: Methodology, Investigation. BGG: Conceptualization, Investigation. JJA: Conceptualization, Validation, Writing – review & editing.

Authors’ information

Not applicable.

Corresponding author

Correspondence to J. J. Arranz.

Ethics declarations

Ethics approval and consent to participate

All procedures involving animals in this study were performed in accordance with Spanish regulations regarding the protection of animals used for experimental and other scientific proposes (Royal Decree 53/2013) under the supervision of the Ethical and Animal Welfare Committee of the University of León after the approval of the competent body, Junta de Castilla y León. The nutritional challenge described in this work was approved by the Ethics Committee of the Instituto de Ganadería de Montaña (IGM, CSIC-ULE) in León (Spain) (Reference 100102/2018–1). In addition, the experimental design and the analysis performed in the current study are in accordance with ARRIVE guidelines.

Consent for publication

Not applicable.

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.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fonseca, P.A.S., Suárez-Vega, A., Esteban-Blanco, C. et al. Epigenetic regulation of functional candidate genes for milk production traits in dairy sheep subjected to protein restriction in the prepubertal stage. BMC Genomics 24, 511 (2023). https://doi.org/10.1186/s12864-023-09611-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12864-023-09611-y

Keywords