Skip to main content
  • Research article
  • Open access
  • Published:

Extracellular vesicle miRNome during subclinical mastitis in dairy cows

Abstract

Bovine mastitis is one of the main inflammatory diseases that can affect the udder during lactation. Somatic cell counts and sometimes microbiological tests are routinely adopted during monitoring diagnostics in dairy herds. However, subclinical mastitis is challenging to identify, reducing the possibility of early treatments. The main aim of this study was to investigate the miRNome profile of extracellular vesicles isolated from milk as potential biomarkers of subclinical mastitis. Milk samples were collected from a total of 60 dairy cows during routine monitoring tests. Small RNA sequencing technology was applied to extracellular vesicles of milk samples collected from cows classified according to the somatic cell count to identify differences in the miRNome between mastitic and healthy cows. A total of 1997 miRNAs were differentially expressed between both groups. Among them, 68 miRNAs whose FDRs were < 0.05 were mostly downregulated, with only one upregulated miRNA (i.e., miR-361). Functional analysis revealed that miR-455-3p, miR-503-3p, miR-1301-3p and miR-361-5p are involved in the regulation of several biological processes related to mastitis, including immune system-related processes. This study suggests the involvement of extracellular vesicle-derived miRNAs in the regulation of mastitis. Moreover, these findings provide evidence that miRNAs from milk extracellular vesicles can be used to identify biomarkers of mastitis. However, further studies must be conducted to validate these miRNAs, especially for subclinical diagnosis.

Introduction

Bovine mastitis is the principal cause of economic losses in the dairy industry due to milk production and quality reduction [1]. In addition, dairy farmers have to address increased costs for treatments and the increased culling rates caused by mastitis outbreaks [2]. The udder infection is caused mostly by a variety of microorganisms, mainly bacteria (Staphylococcus spp., Streptococcus spp., and Enterobacteriaceae), but also by yeast belonging to Candida spp. or protozoa of the genus Prototheca [3]. The clinical classification of mastitis differentiates the disease into clinical and subclinical forms according to the presence or absence of symptoms and signs, such as visibly abnormal milk, swelling, heat, pain and redness of the udder [3]. Subclinical forms are the main challenge for mastitis control due to the normal presentation of both the udder and milk, but with increased somatic cell count (SCC) and the presence of bacteria in milk [4]. According to recent literature on the prevalence of bovine mastitis worldwide, subclinical mastitis is the most prevalent and causes major economic losses, with an estimated value of 2 billion USD per year [5,6,7]. In particular, in Europe, the prevalence of subclinical mastitis is 37% [5], whereas in Italy, it is 22.5% [8]. Therefore, the early identification of subclinical forms is fundamental for adequate treatment and sustainable dairy herd management. Furthermore, with Regulation (EU) 2019/6 of the European Parliament about veterinary medicinal products, the European Union introduced strict limitations on the use of antimicrobials for prophylaxis and metaphylaxis purposes to control the rise and spread of resistance phenomena. In the past, the preventive use of antimicrobials in dairy herds was frequently adopted, especially during the dry period [1, 9]. In the new regulatory scenario, veterinary practitioners working in the dairy industry require alternative solutions for the control of mastitis, including genomic selection for resistance [10] and novel diagnostic and therapeutic tools [1].

Monitoring subclinical mastitis is an essential requirement in dairy herd management and consists of SCC measurements and microbiological tests [3]. Moreover, the SCC parameter is also considered to define milk suitability for human consumption and quality for cheese processing; therefore, milk pricing strictly depends on the SCC [9]. Unanimous agreement considers milk with an SCC value greater than 400 000 cells/mL, regardless of the presence of clinical symptoms, as mastitic [2, 11]. On the other hand, milk from a healthy udder has an SCC value lower than 100 000 cells/mL [2, 9]. An SCC measurement between those values should be interpreted: it could be typical of subclinical mastitis or a finding related to the recovery processes of the mammary gland after infection, when inflammation and pathogens could still be found [11, 12]. Currently, an SCC value equal to 200 000 cells/mL has been adopted as a threshold to diagnose subclinical mastitis [13]. However, inflammation of the mammary gland has also been observed at values of 100 000 cells/mL, especially in primiparous cows [14]. Thus, new biomarkers would help to discriminate cows with subclinical mastitis.

In recent years, extracellular vesicles (EVs) have been widely investigated in human and veterinary medicine. EVs are membrane-limited nanoparticles involved in intercellular communication, and their presence has been proven in several biological fluids, such as blood, urine, milk, and saliva [15,16,17].

Among EVs, three main classes are included: exosomes, ectosomes and apoptotic bodies [18]. Classes are defined according to the range size of EVs and are divided into small-sized EVs (<200 nm) and medium/large-sized EVs (>200 nm) [19]. Owing to their biogenesis, exosomes can be generated through the endosomal complex required for transport (ESCRT) or the ceramide-dependent pathway. The exosome size is normally lower than 100 nm [20]. In contrast, ectosomes originate through exocytosis from the cell membrane. In this case, the dimensions are larger than those of exosomes, and their size can reach 1000 nm [21]. Finally, the classification of apoptotic bodies into EVs is still under debate, and they are generated during apoptosis and are highly variable in size [19].

The ability of EVs to regulate cellular and organ processes is due to the presence of different types of cargo inside EVs, such as noncoding RNA, mRNA, DNA, proteins and lipids, which can be delivered to the targeted recipient cell [20]. The ability of EVs to regulate cellular communication also relies on the presence of several protein and glycoprote in membrane markers [22]. In the last decade, EVs have been largely studied and evaluated as innovative biomarkers in human medicine, particularly for cancer and neurodegenerative diseases.

In veterinary medicine, the role of microRNAs (miRNAs) has been particularly investigated in mastitis pathogenesis among domesticated ruminant species (i.e., cows, sheep and goats). miRNAs are an extremely important group of small noncoding RNAs, ranging in length from 20 to 22 nucleotides, that regulate gene expression through mRNA silencing at the posttranscriptional level [23]. Several studies have been conducted in experimentally infected cows, and several putative miRNAs that are differentially regulated, mainly under Staphylococcus aureus or Escherichia coli infection, have been identified [24,25,26]. Similar to other infectious diseases, during mastitis, miRNAs are involved in the regulation of several pathways, such as pathogen response, activation and regulation of inflammatory processes and regulation of the immune response [27]. The identification of early indicators for rapid and accurate detection of mastitis could lead to earlier and more effective treatment, allowing the animal to recover faster and therefore reducing the associated economic losses. In addition, a better understanding of the molecular regulation of the mammary response to inflammation would allow the identification of such robust indicators. In this context, the main aims of this study were to investigate the role of miRNAs carried by EVs in the regulation of mastitis and to identify putative biomarkers for the early diagnosis of mastitis.

Materials and methods

Study design and sample collection

As previously described [28], a total of 120 primiparous Holstein Friesian cows belonging to 10 dairy farms located in the provinces of Cuneo and Turin (Piedmont Region, Northern Italy) were selected without any clinical signs of disease. Animals were managed according to local farm production practices. All manipulations were performed according to the best animal handling and veterinary practices to avoid animal distress. Before sample collection, the teats were disinfected, and the first milk ejected was collected in a specific container and then discarded. Individual samples were collected in sterile polypropylene tubes (2 aliquots of 50 mL) from all quarters of each cow and immediately stored at 4 °C. Before EV isolation and sequencing, a total of 60 milk samples were randomly selected and further processed for EV isolation. Milk sampling was conducted during the winter (December 2020–February 2021, n = 52) and summer (June 2021–September 2021, n = 8) seasons. Before sequencing, samples were clustered using an SCC threshold level of 200 000 cells/mL, normally considered the SCC level of subclinical mastitis, into two groups: high SCC (group H, n = 11) and low SCC (group L, n = 49). The milk samples in the two groups are shown in Table 1.

Table 1 Milk parameters of the high (H) and low (L) somatic cell count (SCC) groups

EVs isolation

Milk aliquots were processed with consecutive centrifugations, whose steps were as follows: 3000 × g for 10 min at 4 °C to remove milk fat and somatic cells, an additional step of 3000 × g for 10 min at 4 °C to remove additional fat and cell residuals, 5000 × g for 30 min at 4 °C to remove larger cell debris and finally 10 000 × g for 30 min at 4 °C to remove smaller cell debris. After these centrifugation steps, the skim milk was stored at −80 °C until further analyses.

Then, the skim milk samples were thawed gradually on ice. EVs isolation was performed by size exclusion chromatography (SEC), in detail, with the original 70 mm columns of qEV (Izon Science, Lyon, France). Before loading milk samples on the SEC column, the starting volume of 4 mL was reduced using a centrifugation filter tube (AMICON ULTRA-4 50 kDa, Merck Millipore, Burlington, MA, USA) to reach a volume of 500 µL, the maximal loadable capacity of the SEC column. Centrifugation was performed at 4000 × g at 4 °C for 40 min to 60 min, depending on the viscosity of the sample. To avoid protein precipitation at the filter level, during centrifugation, samples were gently mixed by slow pipetting on ice every 5–10 min pausing the centrifugation. Once the volume reached 500 µL, milk samples were loaded on the SEC column, which was previously mounted on its automatic fraction collector (qEV AFC system, Izon Science). Following the manufacturer’s instructions, the first five aliquots (50 µL each) were separately collected in 2 mL tubes corresponding to the most rich and pure EVs fractions. The five EVs fractions (250 µL) were subsequently mixed, and the volume was reduced to reach 50 µL, the minimal useful volume for downstream applications, via a centrifugation filter tube (AMICON ULTRA-4 50 kDa, Merck Millipore). Centrifugation was performed at 4000 × g at 4 °C with a variable time from 15 min to 30 min depending on the sample viscosity. To avoid protein precipitation at the filter level, during centrifugation, samples were gently mixed by slow pipetting on ice every 5–10 min pausing the centrifugation. Finally, the concentrated EVs were stored at -80 °C until further analyses.

EVs validation

Nanoparticle tracking analysis (NTA)

EV concentrations were calculated via nanoparticle tracking analysis (NTA) using the Nanosight LS300 system (Malvern Panalytical, Malvern, UK) equipped with a 488 nm laser. EVs were diluted (1:200) in 0.1 μm filtered saline solution and analysed via NTA 3.2 Analytical Software. Three videos of 60 s at camera level 15 and threshold 5 were captured via a syringe pump 50. The settings were kept constant between samples.

Transmission electron microscopy (TEM)

Transmission electron microscopy (TEM) was performed to evaluate EV integrity and size. EVs were fixed on 200 mesh nickel formvar carbon-coated grids (Electron Microscopy Science, Hatfield, PA, USA) as reported by Bruno et al. [29]. EVs were negatively stained using NanoVan™ and Nano-W™ (Nanoprobes, Yaphank, NY, USA) and observed using a Jeol JEM 1010 electron microscope (Jeol, Tokyo, Japan).

Western blot

To validate the EV isolation method and confirm the presence of EVs in the samples, western blot analysis was conducted. In particular, three EV markers were selected as suggested by the MISEV guidelines: TSG101, CD9 and calnexin [19]. Total proteins were extracted from EV lysates using RIPA buffer supplemented with a protease inhibitor cocktail (Sigma‒Aldrich, St. Louis, MO, USA) and quantified via a DC protein assay (Bio-Rad, Hercules, CA, USA). Equal amounts of protein (300 µg/lane) were resolved with 4–20% MP TGX Stain-Free Gel (Bio-Rad) under reducing conditions for only TSG101 and Calnexin. Proteins were blotted onto PVDF membranes (Trans-Blot Turbo Mini PVDF, Bio-Rad) using Mini Trans-Blot cells (Bio-Rad). The blotted membranes were blocked with a 10% BSA solution (Merck Millipore) for 1 h at room temperature, followed by an overnight incubation at 4 °C with a mouse monoclonal anti-TSG101 primary antibody (1:200; sc-7964, Santa Cruz Biotechnology, Dallas, TX, USA), a mouse monoclonal anti-CD9 primary antibody (1:500; MCA469GT, Bio-Rad) and a rabbit polyclonal anti-calnexin antibody (1:200, sc-11397, Santa Cruz Biotechnology). The membranes were subsequently incubated with secondary horseradish peroxidase (HRP)-conjugated anti-mouse (1:10 000) and anti-rabbit (1:40 000) antibodies and developed with Clarity Western ECL Substrate (Bio-Rad). The immunoblot bands were visualized via the ChemiDoc MP System (Bio-Rad). For western blot analysis, a ready-to-use protein extract from A-431 cell lysate (sc-2201, Santa Cruz Biotechnology) was used as a positive control for calnexin.

Small RNAs extraction

Total small RNAs were extracted from EV samples via a Maxwell RSC miRNA Blood Kit (Promega, Madison, WI, USA) following the manufacturer’s instructions. The input miRNAs were subsequently quantified using the Qubit microRNA Assay Kit. Finally, the small RNA profiles of the input RNA samples were investigated via the Agilent small RNA kit for the Bioanalyzer 2100 instrument (Agilent Technologies, Santa Clara, CA, USA).

Small RNA sequencing

Library preparation for next-generation sequencing was performed using SMARTer smRNA-seq kit for Illumina (Cat. no. #635031; Clontech Laboratories Inc., Kusatsu, Japan) according to the manufacturer’s protocol. Following PCR amplification, purification, and validation, the size selection of the sequencing libraries was performed using SPRIselect beads (Cat. no. #B23318, Beckman Coulter Life Science, Brea, CA, USA). Quality controls were performed on a Bioanalyzer 2100 (Agilent Technologies) and Qubit V4 (Thermo Fisher Scientific). Next-generation sequencing was performed on a NextSeq500 (Illumina, San Diego, CA, USA) with the reagents kit V2 (75 cycles; Illumina). The samples were processed starting from single-ended 75 bp-long sequencing reads.

Bioinformatic analysis

The FASTQ files were trimmed using cutadapt (cutadapt 3.5 with Python 3.7.7) following the manufacturer’s instructions (parameters: -m 15 -u 3 -a AAAAAAAAAA). Trimmed fastq files were processed using the miRNAseq workflow implemented in Docker4seq [30,31,32]. Briefly, quality control of trimmed reads was performed using the FastQC software v. 0.11.9 [33]. The quality of the trimmed reads was checked to evaluate the overall distribution of sequenced fragment length. The trimmed reads were subsequently mapped onto bovine miRNA precursors (miRbase 22) using the BWA aligner (v. 0.7.12) [34], and mature 5p and 3p miRNAs were counted using an R script embedded in the docker4seq workflow.

Count filtering, data normalization, and differential expression analysis were performed in RStudio. We first normalized the miRNA count matrix with the sequencing depth for each sample by calculating counts per million (CPM). Then, we filtered out genes expressed in fewer than 10 samples with CPM < 0.5 via the cpm() function from the edgeR package (v. 3.36.0) [35]. miRNAs failing these criteria were removed from the count matrix before exploration and differential expression analyses.

Once a filtered miRNA matrix was obtained, exploratory analysis of the expressed miRNAs was performed via unsupervised principal component analysis (PCA) and nonparametric multidimensional similarity (NMDS) analysis with the ggplot2 (v. 3.3.5) R package [36]. Differentially expressed (DE) miRNA analysis was performed pairwise via the edgeR (v. 3.36.0) package [35]. Differential analysis was performed by comparing samples according to their SCC threshold level of 200 000 cells/mL. Counts from expressed genes were first normalized with the calcNormFactors() function [37]. The voom() function from the limma R package (v. 3.50.0) was subsequently used to fit a generalized linear regression model to correct the data with the group as a fixed effect [38]. The p values were adjusted for multiple testing via the Benjamini and Hochberg procedure [39]. Only DE miRNAs with an adjusted P value < 0.05 were used for downstream pathway analysis.

In silico functional analysis

Using only DE miRNAs, a predictive functional analysis was performed to evaluate the influence of the DE miRNAs on biological processes and pathways. In silico analyses were performed using human orthologues (hsa-), instead of bovine orthologues (bta-), since information about miRNA activity is more detailed and extensive in humans than in bovines. In this particular step, we used miRWalk 2.0 [40], OmicsNet 2.0 [41] and Cytoscape v.3.9.1 with the ClueGO plugin v.2.5.9 [42] software. First, the most deposited mature forms (-3p or -5p) of DE miRNAs were checked in the miRBase database and modified accordingly in the DE miRNA list with the aim of retrieving the most relevant results via functional analysis. The partially modified DE miRNA list was subsequently used in miRWalk to retrieve all the miRNA-gene interactions. In particular, only validated interactions deposited in the miRTarBase database and with biding position data (related to the 3’UTR, 5’UTR and CDS) were selected. In parallel, to identify functional biological categories related to DE miRNAs, Panther biological process analysis was performed via OmicsNet. Then, the categorization of biological processes was manually performed. Biological processes were clustered into two main macro-categories: cell life processes (including cell cycle, regulation of the cell cycle, cell proliferation and apoptotic process) and gene expression machinery processes (including regulation of transcription by RNA polymerase II, transcription DNA templating, mRNA processing, mRNA splicing via the spliceosome, and regulation of translation and protein phosphorylation). In addition, target genes involved in the regulation of immune processes were obtained with the ClueGO plugin in Cytoscape via the function GO-ImmuneSystemProcess. These gene lists were filtered by selecting only validated miRNA-gene interactions previously obtained via miRWalk. The miRNAs-genes network was visualized via Cytoscape, which was subsequently used to identify potential networks.

Results

EV validation by NTA, TEM and Western blot analyses

To validate the presence of EVs in the samples, NTA revealed an average size distribution of EVs of 171.87 ± 33.93 nm (Figure 1A), and TEM analysis (Figure 1B) confirmed that the EVs had a homogeneous pattern of nanosized membrane vesicles. Moreover, western blot analysis was performed on representative samples randomly selected from the collected samples. Both EV markers, TSG101 and CD9, were positive in the samples analysed, whereas calnexin was negative in the EV samples. The specific bands of the two targets are reported in Figure 1C.

Figure 1
figure 1

EV validation. Representative graphs of the results of nanoparticle tracking analysis (NTA) showing EV size distribution (A). Representative TEM images of EVs showing intact and heterogeneous EVs; top and bottom scale bars, 200 nm and 100 nm, respectively (B). Western blot analysis of EV samples. 1: isotype control antibody on EVs, 2: EVs, 3: cell lysate (A-431 cell line) (C). The EV markers used for analysis were TSG-101 (45 kDa), CD9 (24 kDa) and calnexin (90 kDa).

Differential analyses of miRNAs

In total, 2127 Bos taurus-annotated miRNAs (bta-miRNAs) were detected. A PCA table was generated to investigate the sample distribution (Additional file 1). Using a cut-off value for the SCC parameter of 200 000 cells/mL, a total of 68 miRNAs were differentially expressed (DE) with a false discovery rate (FDR) < 0.05 between group H and group L (miRNAs were up-/downregulated in group H in comparison to group L). These genes were mostly downregulated. Intriguingly, only 1 miRNA, bta-miR-361-3p, was upregulated, with a 2.7-fold change (FC) value. Among the downregulated miRNAs, 15 had a FC < −2. A volcano plot of the DE miRNAs is shown in Figure 2. The list of the 68 DE miRNAs is reported in Additional file 2.

Figure 2
figure 2

Differentially expressed miRNAs between group H and group L. The regulation of expression is intended to be downregulated/upregulated in group H in comparison with that in group L.

Functional analysis of DE miRNAs

The functional analysis was conducted using human orthologues (hsa-) instead of bovine orthologues (bta-) since human databases are more complete and more informative regarding miRNA function. The targets of 17 known miRNAs were obtained from OmicsNet via the Panther database. In addition, the regulation of immune system processes was investigated via the ClueGO plugin in Cytoscape. The main immune processes significantly influenced by the DE miRNAs were related mainly to mediated immunity, including immunoglobulin production and lymphocyte regulation. Therefore, the miRNA-gene networks were visualized via Cytoscape, and three separate reactomes were generated considering cell life (Figure 2), gene expression (Figure 3) and immune processes (Figure 4). Two large nodes involved in the regulation of miRNA-gene interactions are miR-503-5p and miR-455-3p, which are involved in both cell life and gene expression processes. The third network related to immunity is less complex than the other two networks are. However, 4 main highlighted miRNAs (miR-455-3p, miR-503-3p, miR-1301-3p and miR-361-5p) were involved in the regulation of immunity (Figure 5).

Figure 3
figure 3

Networks of the differentially abundant miRNAs identified via Cytoscape. These results are related to miRNA-gene interactions involved in cell life processes.

Figure 4
figure 4

Networks of the differentially abundant miRNAs identified via Cytoscape. These results are related to the miRNA-gene interactions involved in the gene expression machinery.

Figure 5
figure 5

Networks of the differentially abundant miRNAs identified via Cytoscape. These results are related to miRNA-gene interactions involved in immune system processes.

Discussion

Bovine mastitis is one of the main challenges that farmers and veterinarians routinely face in dairy farming. Moreover, the spread of antimicrobial resistance and the recent enactment of the new European Regulation about the use of veterinary medicinal products have increased the need for the implementation of diagnostic tools to detect subclinical mastitis early to reduce/avoid antimicrobial treatments [43]. Among the different parameters of mammary gland infection, an increase in the SCC in milk is considered to have prognostic value. Therefore, to compare healthy subjects with cows potentially affected by subclinical mastitis, the milk samples were classified according to the SCC measurement, setting the 200 000 cell/mL value as a threshold [9]. In this study, the miRNome profile of dairy cows affected by subclinical mastitis was investigated and compared with that of healthy control samples. EVs were obtained from milk samples to better understand the role of miRNAs in the regulation of mastitis and to identify new potential biomarkers of the disease. EVs and miRNAs have been largely studied in human and veterinary medicine for their evaluation as potential biomarkers of different diseases, such as cancer and neurodegenerative and infectious diseases. In this study, individual milk samples were collected from a total of 60 Holstein Friesian cows during routine mastitis screening tests and subjected to EV small RNA-seq analysis to detect miRNome profiles suggestive of the subclinical form of the disease. Indeed, bovine mastitis can modify the milk miRNome, and several miRNAs have been reported to be differentially expressed during mammary gland inflammation [23].

The results of the EV miRNome profiling revealed statistically significant differences between samples with high SCC levels (>200 000 cell/mL, group H, considered a group with subclinical mastitis) and samples with low SCC levels (<200 000 cell/mL, group L, considered a healthy control group). In particular, 68 miRNAs were DE, most of which were downregulated in group H compared with group L. Among these 68 DE miRNAs, 15 miRNAs were downregulated, with FC values < -2. Many of these DE miRNAs were specifically related to bovine species, and no relevant information could be retrieved from the scientific literature. However, three miRNAs can be highlighted: miR-223, miR-124 and miR-568. The first one, i.e., miR-223, is largely described in the literature as regulating the inflammatory response in bovine mastitis [44,45,46,47,48]. All these studies reported the upregulation of miR-223 in response to S. aureus and S. uberis. or S. agalactiae experimental infections. Therefore, the finding of miR-223 downregulation in the present study is not consistent with what has been reported in the scientific literature. One possible explanation for this controversial result may be related to the different matrices used for miRNA extraction. The abovementioned studies investigated the role of miRNAs in different mastitis models involving mammary gland biopsies [44, 46, 47], cultured Mac-T cells [24] or blood [45]. Therefore, an adequate comparison is not possible. Moreover, in the dataset of this study, only two samples were positive for S. uberis, which may not be enough to highlight differences between the groups. In contrast, Tzelos and colleagues investigated the expression of miR-223 during mastitis, but a statistically significant difference in expression was not detected between mastitic and healthy cows [49]. In addition, in Tzelos’s study, miR-223 was investigated both in whole and skim milk samples and not in milk EVs, affecting the possibility of an adequate comparison. However, according to the human medical literature, miR-223 is either expressed almost exclusively or highly enriched in several subsets of white blood cells, including T- and B-lymphocytes, neutrophils, mast cells and monocytes [50]. It is well known that miRNAs that can be detected in milk can have different origins according to the different cells composing and characterizing the mammary gland during mastitis, i.e., mammary epithelial cells, inflammatory cells, adipocytes, fibroblasts or myoepithelial cells [36]. On the other hand, miR-124 and miR-568 were also significantly downregulated in this study and, for the first time, were reported to be related to bovine mastitis. According to the literature, miR-124 has been described to regulate the parasitic pathogenesis of Schistosoma japonicum and Fasciola gigantica in both bovines and buffaloes [51,52,53]. Intriguingly, the inhibitory effect of miR-568 on CD4+ T cells was previously demonstrated, suggesting a role in the modulation of lymphocyte activity [54].

According to the functional analysis, four miRNAs (namely, miR-455, miR-361, miR-1301, and miR-503) were found to be involved in the regulation of the biological processes of gene expression, cell life and the immune response. As mentioned previously, the functional analysis was conducted using human orthologues (hsa-) rather than bovine orthologues (bta-). Given this possible bias, the functional results obtained in this study may be considered predictive, and further studies must be conducted to validate these predictions. However, the results can be compared with the scientific literature to contextualize and explain the roles of these four miRNAs. MiR-455 has already been reported in dairy cows subjected to dietary regulation. For example, Webb and colleagues reported a downregulation of miR-455 in the period after calving in cows fed a highly balanced diet [55]. Our results are consistent with those of this study since both the downregulation of miR-455 and its involvement in the regulation of the inflammatory response have been reported. According to the literature in humans, miR-455 downregulation is associated with the activation of inflammatory pathways in multiple sclerosis [56]. Moreover, the anti-inflammatory activity of miR-455 was also described by recent studies reporting an efficient therapeutic role of this miRNA in acute liver injury and cerebral ischemia/reperfusion injury [57, 58]. Another interesting result is miR-361 upregulation, which seems to be involved in the regulation of biological processes related to the immune response. In bovine, the level of miR-361 has been reported to be lower in the serum of grazing cows than in that of housed cattle [59]. This study may suggest a role for miR-361 in the regulation of metabolism in cows. It is well known that mastitis affects cow metabolism by altering lipolysis and the milk proteome [60, 61]. Therefore, the upregulation of miR-361 in our samples may depend on metabolic factors modified by mastitis. In addition, in humans, miR-361 is reported to be a promising biomarker for tuberculosis diagnosis and is most likely involved in the regulation of host‒pathogen interactions [62, 63]. The downregulation of miR-1301 was first reported in mastitis in this study. Luoreng and colleagues reported the upregulation of miR-1301 in the blood of dairy cows experimentally infected with a Staphylococcus aureus strain [64]. This controversial result may be partially explained by the different biological fluids analysed, the milk used in the present study and the blood used in the previous study. Furthermore, S. aureus was never identified in the milk samples of this study. It is well known that etiological agents can influence mastitis pathogenesis differently [26, 64], and differences in the milk miRNome between mastitis caused by either Escherichia coli or S. aureus have already been reported [65]. Therefore, the differential regulation of miR-1301 depending on the specific pathogen could not be excluded. Finally, miR-503 represents another important node of regulation according to the functional analysis. Most information regarding miR-503 is related to human diseases. This miRNA seems to be involved in the pathogenesis of diabetes and lipopolysaccharide injury [66, 67], but more intriguingly, its downregulation has been reported to be involved in the activation of NF-kB signalling and the PPARγ pathway [68, 69]. In veterinary medicine, the downregulation of miR-503 has also been reported in dog blood mononuclear cells infected with Leishmania infantum, suggesting a role in the modulation of the host‒response [70].

One innovative aspect of this study is the selection of dairy cows naturally affected by mastitis. Studies with an in-field scenario allow the evaluation of pathological conditions while taking into account the actual environmental variability. Moreover, some studies, even if conducted on naturally infected cows, considered a very low sample size [71, 72] in comparison with the sample size in this study (60 Holstein Friesian cows). Considering the method applied, another important consideration can be made. Small RNA-seq allows the investigation of the wide and complete panorama of miRNA expression and activity. On the other hand, studies applying qPCR or microarrays can focus only on selected and limited miRNAs and do not allow the identification of novel bovine miRNAs [49, 73, 74]. Finally, the other two studies, from Bagnicka et al. and Özdemir, represent the most comparable studies with the present study [75, 76]. Similar methods for small RNA-seq and a smaller (but still well representative) sample size have been applied. However, the main DE miRNAs reported are different from those identified in the present study. The main reason is likely related to the animal selection modality, which analysed only dairy cows positive for coagulase +/-Staphylococci or M. bovis. Instead, the present study focused on the selection of cows on SCC and not on bacteriological results. Furthermore, as already mentioned, the different biological matrices could influence the results. In fact, the origin of miRNAs can affect the miRNA profile. For example, whole milk has a different miRNA profile than skim milk, somatic cells, milk fat globules, or mammary gland biopsies [77,78,79].

In conclusion, the results obtained from this study could be a promising starting point for the investigation of miRNA and EV regulation in bovine subclinical mastitis. The functional analysis revealed a panel of four miRNAs that are promising putative biomarkers of subclinical mastitis: miR-455, miR-361, miR-1301 and miR-503. Notably, these results were validated via predictive analysis using human orthologues; therefore, their role may not be the same in bovines. To clarify the DE miRNA results, future validation via qPCR or ddPCR may be performed on other dairy cows. Furthermore, to define the role of DE miRNAs in the regulation of genes identified by functional analysis and therefore in inflammation, an in vitro luciferase reporter gene assay to validate miRNA target sites in bovines may also be performed. Nevertheless, the poor presence of mastidogen bacteria observed in the dataset may represent a limitation of this study, and an additional validation of the obtained results may involve the inclusion of gram-negative or Mycoplasma spp. mastitis forms. Further studies must be conducted to identify and validate miRNAs used for mastitis detection, especially subclinical forms. These methods can be considered integrative approaches among clinical evaluation, SCC, microbiology and miRNAs, which will always constitute the optimal strategy for early detection of bovine mastitis.

Availability of data and materials

The dataset(s) supporting the conclusions of this article is (are) available in the Genome Sequence Archive (GSA) repository, identified with the code subCRA018425 at https://ngdc.cncb.ac.cn/gsub/submit/biosample/list.

References

  1. El-Sayed A, Kamel M (2021) Bovine mastitis prevention and control in the post-antibiotic era. Trop Anim Health Prod 53:236

    Article  PubMed  Google Scholar 

  2. Ruegg PL (2017) A 100-Year review: Mastitis detection, management, and prevention. J Dairy Sci 100:10381–10397

    Article  CAS  PubMed  Google Scholar 

  3. Ashraf A, Imran M (2020) Causes, types, etiological agents, prevalence, diagnosis, treatment, prevention, effects on human health and future aspects of bovine mastitis. Anim Health Res Rev 21:36–49

    Article  PubMed  Google Scholar 

  4. Ashraf A, Imran M (2018) Diagnosis of bovine mastitis: from laboratory to farm. Trop Anim Health Prod 50:1193–1202

    Article  PubMed  Google Scholar 

  5. Krishnamoorthy P, Goudar AL, Suresh KP, Roy P (2021) Global and countrywide prevalence of subclinical and clinical mastitis in dairy cattle and buffaloes by systematic review and meta-analysis. Res Vet Sci 136:561–586

    Article  PubMed  Google Scholar 

  6. Krömker V, Leimbach S (2017) Mastitis treatment-reduction in antibiotic usage in dairy cows. Reprod Domest Anim Zuchthyg 52(Suppl 3):21–29

    Article  Google Scholar 

  7. Moroni P, Pisoni G, Antonini M, Villa R, Boettcher P, Carli S (2006) Short communication: antimicrobial drug susceptibility of Staphylococcus aureus from subclinical bovine mastitis in Italy. J Dairy Sci 89:2973–2976

    Article  CAS  PubMed  Google Scholar 

  8. Zecconi A, Vairani D, Cipolla M, Rizzi N, Zanini L (2019) Assessment of subclinical mastitis diagnostic accuracy by differential cell count in individual cow milk. Ital J Anim Sci 18:460–465

    Article  CAS  Google Scholar 

  9. National Mastitis Council (2001) Guidelines on normal and abnormal raw milk based on somatic cell counts and signs of clinical mastitis

  10. Moretti R, Chessa S, Sartore S, Soglia D, Giaccone D, Cannizzo FT, Sacchi P (2022) A practical application of genomic predictions for mastitis resistance in Italian holstein heifers. Animals 12:2370

    Article  PubMed  PubMed Central  Google Scholar 

  11. Zecconi A, Meroni G, Sora V, Mattina R, Cipolla M, Zanini L (2021) Total and differential cell counts as a tool to identify intramammary infections in cows after calving. Animals 11:727

    Article  PubMed  PubMed Central  Google Scholar 

  12. Adkins PRF, Middleton JR (2018) Methods for diagnosing mastitis. Vet Clin North Am Food Anim Pract 34:479–491

    Article  PubMed  Google Scholar 

  13. Dufour S, Dohoo IR (2013) Monitoring herd incidence of intramammary infection in lactating cows using repeated longitudinal somatic cell count measurements. J Dairy Sci 96:1568–1580

    Article  CAS  PubMed  Google Scholar 

  14. Kandeel SA, Megahed AA, Arnaout FK, Constable PD (2018) Evaluation and comparison of 2 on-farm tests for estimating somatic cell count in quarter milk samples from lactating dairy cattle. J Vet Intern Med 32:506–515

    Article  CAS  PubMed  Google Scholar 

  15. Yu D, Li Y, Wang M, Gu J, Xu W, Cai H, Fang X, Zhang X (2022) Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer 21:56

    Article  PubMed  PubMed Central  Google Scholar 

  16. Benmoussa A, Michel S, Gilbert C, Provost P (2020) Isolating multiple extracellular vesicles subsets, including exosomes and membrane vesicles, from bovine milk using sodium citrate and differential ultracentrifugation. Bio-Protocol 10:e3636

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Blans K, Hansen MS, Sørensen LV, Hvam ML, Howard KA, Möller A, Wiking L, Larsen LB, Rasmussen JT (2017) Pellet-free isolation of human and bovine milk extracellular vesicles by size-exclusion chromatography. J Extracell Vesicles 6:1294340

    Article  PubMed  PubMed Central  Google Scholar 

  18. Dang XTT, Kavishka JM, Zhang DX, Pirisinu M, Le MTN (2020) Extracellular vesicles as an efficient and versatile system for drug delivery. Cells 9:2191

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, Ayre DC, Bach JM, Bachurski D, Baharvand H, Balaj L, Baldacchino S, Bauer NN, Baxter AA, Bebawy M, Beckham C, Bedina Zavec A, Benmoussa A, Berardi AC, Bergese P, Bielska E, Blenkiron C, Bobis-Wozowicz S, Boilard E, Boireau W, Bongiovanni A et al (2018) Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 7:1535750

    Article  PubMed  PubMed Central  Google Scholar 

  20. Zempleni J, Aguilar-Lozano A, Sadri M, Sukreet S, Manca S, Wu D, Zhou F, Mutai E (2017) Biological activities of extracellular vesicles and their cargos from bovine and human milk in humans and implications for infants. J Nutr 147:3–10

    Article  CAS  PubMed  Google Scholar 

  21. Raposo G, Stoorvogel W (2013) Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 200:373–383

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Admyre C, Johansson SM, Qazi KR, Filén JJ, Lahesmaa R, Norman M, Neve EP, Scheynius A, Gabrielsson S (2007) Exosomes with immune modulatory features are present in human breast milk. J Immunol 179:1969–1978

    Article  CAS  PubMed  Google Scholar 

  23. Do DN, Dudemaine PL, Mathur M, Suravajhala P, Zhao X, Ibeagha-Awemu EM (2021) miRNA regulatory functions in farm animal diseases, and biomarker potentials for effective therapies. Int J Mol Sci 22:3080

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cai M, Fan W, Li X, Sun H, Dai L, Lei D, Dai Y, Liao Y (2021) The regulation of Staphylococcus aureus-induced inflammatory responses in bovine mammary epithelial cells. Front Vet Sci 8:683886

    Article  PubMed  PubMed Central  Google Scholar 

  25. Chen Y, Jing H, Chen M, Liang W, Yang J, Deng G, Guo M (2021) Transcriptional profiling of exosomes derived from Staphylococcus aureus-infected bovine mammary epithelial cell line MAC-T by RNA-seq analysis. Oxid Med Cell Longev 2021:8460355

    Article  PubMed  PubMed Central  Google Scholar 

  26. Jin W, Ibeagha-Awemu EM, Liang G, Beaudoin F, Zhao X, Guan le L (2014) Transcriptome microRNA profiling of bovine mammary epithelial cells challenged with Escherichia coli or Staphylococcus aureus bacteria reveals pathogen directed microRNA expression profiles. BMC Genomics 15:181

    Article  PubMed  PubMed Central  Google Scholar 

  27. Lawless N, Vegh P, O’Farrelly C, Lynn DJ (2014) The role of microRNAs in bovine infection and immunity. Front Immunol 5:611

    Article  PubMed  PubMed Central  Google Scholar 

  28. Cuccato M, Divari S, Sacchi P, Girolami F, Cannizzo FT (2022) MALDI-TOF mass spectrometry profiling of bovine skim milk for subclinical mastitis detection. Front Vet Sci 9:1009928

    Article  PubMed  PubMed Central  Google Scholar 

  29. Bruno S, Chiabotto G, Cedrino M, Ceccotti E, Pasquino C, De Rosa S, Grange C, Tritta S, Camussi G (2022) Extracellular vesicles derived from human liver stem cells attenuate chronic kidney disease development in an in vivo experimental model of renal ischemia and reperfusion injury. Int J Mol Sci 23:1485

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Beccuti M, Cordero F, Arigoni M, Panero R, Amparore EG, Donatelli S, Calogero RA (2018) SeqBox: RNAseq/ChIPseq reproducible analysis on a consumer game computer. Bioinforma Oxf Engl 34:871–872

    Article  CAS  Google Scholar 

  31. Ferrero G, Cordero F, Tarallo S, Arigoni M, Riccardo F, Gallo G, Ronco G, Allasia M, Kulkarni N, Matullo G, Vineis P, Calogero RA, Pardini B, Naccarati A (2018) Small non-coding RNA profiling in human biofluids and surrogate tissues from healthy individuals: description of the diverse and most represented species. Oncotarget 9:3097–3111

    Article  PubMed  Google Scholar 

  32. Kulkarni N, Alessandrì L, Panero R, Arigoni M, Olivero M, Ferrero G, Cordero F, Beccuti M, Calogero RA (2018) Reproducible bioinformatics project: a community for reproducible bioinformatics analysis pipelines. BMC Bioinformatics 19:349

    Article  PubMed  PubMed Central  Google Scholar 

  33. Andrews S (2010) FastQC: a quality control tool for high throughput sequence data. http://www.bioinformatics.babraham.ac.uk/projects/fastqc. Accessed 23 Aug 2024

  34. Li H, Durbin R (2009) Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinforma Oxf Engl 25:1754–1760

    Article  CAS  Google Scholar 

  35. Robinson MD, McCarthy DJ, Smyth GK (2010) edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinforma Oxf Engl 26:139–140

    Article  CAS  Google Scholar 

  36. Wickham H (2016) ggplot2: elegant graphics for data analysis. ggplot2 XVI, p 260

  37. Bullard JH, Purdom E, Hansen KD, Dudoit S (2010) Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments. BMC Bioinformatics 11:94

    Article  PubMed  PubMed Central  Google Scholar 

  38. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK (2015) Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43:e47

    Article  PubMed  PubMed Central  Google Scholar 

  39. Benjamini Y, Hochberg Y (1995) Controlling the false Discovery rate: a practical and powerful Approach to multiple testing. J R Stat Soc Ser B Methodol 57:289–300

    Article  Google Scholar 

  40. Sticht C, De La Torre C, Parveen A, Gretz N (2018) miRWalk: an online resource for prediction of microRNA binding sites. PLoS One 13:e0206239

    Article  PubMed  PubMed Central  Google Scholar 

  41. Zhou G, Pang Z, Lu Y, Ewald J, Xia J (2022) OmicsNet 2.0: a web-based platform for multi-omics integration and network visual analytics. Nucleic Acids Res 50:W527–W533

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, Fridman WH, Pagès F, Trajanoski Z, Galon J (2009) ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 25:1091–1093

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cheng WN, Han SG (2020) Bovine mastitis: risk factors, therapeutic strategies, and alternative treatments — a review. Asian-Australas J Anim Sci 33:1699–1713

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Naeem A, Zhong K, Moisá SJ et al (2012) Bioinformatics analysis of microRNA and putative target genes in bovine mammary tissue infected with Streptococcus uberis J Dairy Sci 95:6397–6408

    Article  CAS  PubMed  Google Scholar 

  45. Chen L, Liu X, Li Z, Wang H, Liu Y, He H, Yang J, Niu F, Wang L, Guo J (2014) Expression differences of miRNAs and genes on NF-κB pathway between the healthy and the mastitis Chinese holstein cows. Gene 545:117–125

    Article  CAS  PubMed  Google Scholar 

  46. Fang L, Hou Y, An J, Li B, Song M, Wang X, Sørensen P, Dong Y, Liu C, Wang Y, Zhu H, Zhang S, Yu Y (2016) Genome-wide transcriptional and post-transcriptional regulation of innate immune and defense responses of bovine mammary gland to Staphylococcus aureus Front Cell Infect Microbiol 6:193

    Article  PubMed  PubMed Central  Google Scholar 

  47. Pu J, Li R, Zhang C, Chen D, Liao X, Zhu Y, Geng X, Ji D, Mao Y, Gong Y, Yang Z (2017) Expression profiles of miRNAs from bovine mammary glands in response to Streptococcus agalactiae-induced mastitis. J Dairy Res 84:300–308

    Article  CAS  PubMed  Google Scholar 

  48. Cai M, He H, Jia X, Chen S, Wang J, Shi Y, Liu B, Xiao W, Lai S (2018) Genome-wide microRNA profiling of bovine milk-derived exosomes infected with Staphylococcus aureus Cell Stress Chaperones 23:663–672

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Tzelos T, Ho W, Charmana VI, Lee S, Donadeu FX (2022) MiRNAs in milk can be used towards early prediction of mammary gland inflammation in cattle. Sci Rep 12:5131

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ludwig N, Leidinger P, Becker K, Backes C, Fehlmann T, Pallasch C, Rheinheimer S, Meder B, Stähler C, Meese E, Keller A (2016) Distribution of miRNA expression across human tissues. Nucleic Acids Res 44:3865–3877

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Guo X, Guo A (2019) Profiling circulating microRNAs in serum of Fasciola gigantica-infected buffalo. Mol Biochem Parasitol 232:111201

    Article  CAS  PubMed  Google Scholar 

  52. Yu X, Zhai Q, Fu Z, Hong Y, Liu J, Li H, Lu K, Zhu C, Lin J, Li G (2019) Comparative analysis of microRNA expression profiles of adult Schistosoma japonicum isolated from water buffalo and yellow cattle. Parasit Vectors 12:196

    Article  PubMed  PubMed Central  Google Scholar 

  53. Zhou X, Hong Y, Shang Z, Abuzeid AMI, Lin J, Li G (2022) The potential role of microRNA-124-3p in growth, development, and reproduction of Schistosoma Japonicum Front Cell Infect Microbiol 12:862496

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lie PPY, Cheng CY, Mruk DD (2013) Signalling pathways regulating the blood-testis barrier. Int J Biochem Cell Biol 45:621–625

    Article  CAS  PubMed  Google Scholar 

  55. Webb LA, Ghaffari MH, Sadri H, Schuh K, Zamarian V, Koch C, Trakooljul N, Wimmers K, Lecchi C, Ceciliani F, Sauerwein H (2020) Profiling of circulating microRNA and pathway analysis in normal- versus over-conditioned dairy cows during the dry period and early lactation. J Dairy Sci 103:9534–9547

    Article  CAS  PubMed  Google Scholar 

  56. Torabi S, Tamaddon M, Asadolahi M, Shokri G, Tavakoli R, Tasharrofi N, Rezaei R, Tavakolpour V, Sazegar H, Kouhkan F (2019) Mir-455-5p downregulation promotes inflammation pathways in the relapse phase of relapsing-remitting multiple sclerosis disease. Immunogenetics 71:87–95

    Article  CAS  PubMed  Google Scholar 

  57. Shao M, Xu Q, Wu Z, Chen Y, Shu Y, Cao X, Chen M, Zhang B, Zhou Y, Yao R, Shi Y, Bu H (2020) Exosomes derived from human umbilical cord mesenchymal stem cells ameliorate IL-6-induced acute liver injury through miR-455-3p. Stem Cell Res Ther 11:37

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang Z, Luo W, Han Y, Misrani A, Chen H, Long C (2022) Effect of microRNA-455-5p (miR-455-5p) on the expression of the cytokine signaling-3 (SOCS3) gene during myocardial infarction. J Biomed Nanotechnol 18:202–210

    Article  CAS  PubMed  Google Scholar 

  59. Muroya S, Ogasawara H, Hojito M (2015) Grazing affects exosomal circulating microRNAs in cattle. PLoS One 10:e0136475

    Article  PubMed  PubMed Central  Google Scholar 

  60. Addis MF, Maffioli EM, Ceciliani F, Tedeschi G, Zamarian V, Tangorra F, Albertini M, Piccinini R, Bronzo V (2020) Influence of subclinical mastitis and intramammary infection by coagulase-negative staphylococci on the cow milk peptidome. J Proteom 226:103885

    Article  CAS  Google Scholar 

  61. Giagu A, Penati M, Traini S, Dore S, Addis MF (2022) Milk proteins as mastitis markers in dairy ruminants—a systematic review. Vet Res Commun 46:329–351

    Article  PubMed  PubMed Central  Google Scholar 

  62. Qi Y, Cui L, Ge Y, Shi Z, Zhao K, Guo X, Yang D, Yu H, Cui L, Shan Y, Zhou M, Wang H, Lu Z (2012) Altered serum microRNAs as biomarkers for the early diagnosis of pulmonary tuberculosis infection. BMC Infect Dis 12:384

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Ndzi EN, Nkenfou CN, Mekue LM, Zentilin L, Tamgue O, Pefura EWY, Kuiaté JR, Giacca M, Ndjolo A (2019) MicroRNA hsa-miR-29a-3p is a plasma biomarker for the differential diagnosis and monitoring of tuberculosis. Tuberculosis 114:69–76

    Article  CAS  PubMed  Google Scholar 

  64. Luoreng ZM, Wang XP, Mei CG, Zan LS (2018) Comparison of microRNA profiles between bovine mammary glands infected with Staphylococcus aureus and Escherichia coli Int J Biol Sci 14:87–99

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mansor R, Mullen W, Albalat A, Zerefos P, Mischak H, Barrett DC, Biggs A, Eckersall PD (2013) A peptidomic approach to biomarker discovery for bovine mastitis. J Proteom 85:89–98

    Article  CAS  Google Scholar 

  66. De Silva N, Samblas M, Martínez JA, Milagro FI (2018) Effects of exosomes from LPS-activated macrophages on adipocyte gene expression, differentiation, and insulin-dependent glucose uptake. J Physiol Biochem 74:559–568

    Article  PubMed  Google Scholar 

  67. Zapała B, Kamińska A, Piwowar M, Paziewska A, Gala-Błądzińska A, Stępień EŁ (2023) miRNA signature of urine extracellular vesicles shows the involvement of inflammatory and apoptotic processes in diabetic chronic kidney disease. Pharm Res 40:817–832

    Article  PubMed  PubMed Central  Google Scholar 

  68. Lee A, Papangeli I, Park Y, Jeong HN, Choi J, Kang H, Jo HN, Kim J, Chun HJ (2017) A PPARγ-dependent miR-424/503-CD40 axis regulates inflammation mediated angiogenesis. Sci Rep 7:2528

    Article  PubMed  PubMed Central  Google Scholar 

  69. Zhou R, Gong AY, Chen D, Miller RE, Eischeid AN, Chen XM (2013) Histone deacetylases and NF-kB signaling coordinate expression of CX3CL1 in epithelial cells in response to microbial challenge by suppressing miR-424 and miR-503. PLoS One 8:e65153

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Soares MF, Melo LM, Bragato JP, Furlan AO, Scaramele NF, Lopes FL, Lima VMF (2021) Differential expression of miRNAs in canine peripheral blood mononuclear cells (PBMC) exposed to Leishmania infantum in vitro. Res Vet Sci 134:58–63

    Article  CAS  PubMed  Google Scholar 

  71. Ju Z, Jiang Q, Liu G, Wang X, Luo G, Zhang Y, Zhang J, Zhong J, Huang J (2018) Solexa sequencing and custom microRNA chip reveal repertoire of microRNAs in mammary gland of bovine suffering from natural infectious mastitis. Anim Genet 49:3–18

    Article  CAS  PubMed  Google Scholar 

  72. Saenz-de-Juano MD, Silvestrelli G, Weber A, Röhrig C, Schmelcher M, Ulbrich SE (2022) Inflammatory response of primary cultured bovine mammary epithelial cells to Staphylococcus aureus extracellular vesicles. Biology 11:415

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Lai YC, Fujikawa T, Maemura T, Ando T, Kitahara G, Endo Y, Yamato O, Koiwa M, Kubota C, Miura N (2017) Inflammation-related microRNA expression level in the bovine milk is affected by mastitis. PLoS One 12:e0177182

    Article  PubMed  PubMed Central  Google Scholar 

  74. Srikok S, Patchanee P, Boonyayatra S, Chuammitri P (2020) Potential role of MicroRNA as a diagnostic tool in the detection of bovine mastitis. Prev Vet Med 182:105101

    Article  PubMed  Google Scholar 

  75. Bagnicka E, Kawecka-Grochocka E, Pawlina-Tyszko K, Zalewska M, Kapusta A, Kościuczuk E, Marczak S, Ząbek T (2021) MicroRNA expression profile in bovine mammary gland parenchyma infected by coagulase-positive or coagulase-negative staphylococci. Vet Res 52:41

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Özdemir S (2020) Identification and comparison of exosomal microRNAs in the milk and colostrum of two different cow breeds. Gene 743:144609

    Article  PubMed  Google Scholar 

  77. Li Y, Zang H, Zhang X, Huang G (2020) Exosomal Circ-ZNF652 promotes cell proliferation, migration, invasion and glycolysis in hepatocellular carcinoma via MiR-29a-3p/gucd1 axis. Cancer Manag Res 12:7739–7751

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Pawlowski K, Lago-Novais D, Bevilacqua C, Mobuchon L, Crapart N, Faulconnier Y, Boby C, Carvalho G, Martin P, Leroux C (2020) Different miRNA contents between mammary epithelial cells and milk fat globules: a random or a targeted process? Mol Biol Rep 47:8259–8264

    Article  CAS  PubMed  Google Scholar 

  79. Leroux C, Pawlowski K, Billa P-A, Pires JAA, Faulconnier Y (2022) Milk fat globules as a source of microRNAs for mastitis detection. Livest Sci 263:104997

    Article  Google Scholar 

Download references

Acknowledgements

The authors are grateful to Dr Alessia Finotto, Dr Andrea Salaroglio, Prof. Flavia Girolami and Dr Daniele Giaccone (A.R.A.P.) for their technical assistance with milk sampling, bacteriological analyses and western blotting. Moreover, special acknowledgement is given to all dairy farmers involved in the TECH4MILK project for their availability and support in the project.

Funding

This research was funded by Regione Piemonte (Italy) for the project “Tech4Milk: Tecnologie e soluzioni innovative al servizio della filiera latte piemontese per promuoverne la competitività e sostenibilità” (FESR 2014–2020 – D24I19000980002). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

FTC and PS conceived and designed the study and revised and edited the final version of the manuscript. MC, SD, CG and AR performed the analyses. DG and MC performed the bioinformatic analyses. DG, RM, CL and FTC supervised the bioinformatic analyses. MC, SD, DG and CL performed data curation and statistical analysis. FTC and PS supervised the study. FTC and PS acquired the funding. MC wrote the original draft. SD, DG, CG, RM, CL, PS, and FTC reviewed and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Sara Divari.

Ethics declarations

Ethics approval and consent to participate

Ethical review and approval were not required for the animal study because milk was collected during the routine monitoring of milk quality on farms. Animals were managed according to local farm-production practices. All manipulations were performed kindly to avoid animal distress. Written informed consent was obtained from the owners for the participation of their animals in this study.

Competing interests

The authors declare that they have no competing interests.

Additional information

Communicated by Tom McNeilly.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: PCA table for sequenced miRNAs.

Additional file 2: 

DE miRNA list. In Additional file 1, the list of DE miRNAs with related FC and p value data is reported, with an SCC cut-off of 200,000 cells/mL used for differential analysis.

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

Cuccato, M., Divari, S., Giannuzzi, D. et al. Extracellular vesicle miRNome during subclinical mastitis in dairy cows. Vet Res 55, 112 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13567-024-01367-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13567-024-01367-x

Keywords