RESEARCH ARTICLE

Comparative miRNome analysis of colostrum- and mature milk–derived extracellular vesicles from Holstein and Jersey cows

Daye Mun1,#https://orcid.org/0000-0002-3470-7632, Sangdon Ryu2,#https://orcid.org/0000-0001-5338-8385, Dong-Hyun Lim3https://orcid.org/0000-0002-8575-0198, Sangnam Oh4https://orcid.org/0000-0002-2428-412X, Younghoon Kim1,*https://orcid.org/0000-0001-6769-0657
Author Information & Copyright
1Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
2Honam National Institute of Biological Resources, Mokpo 58762, Korea
3Dairy Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Korea
4Department of Functional Food and Biotechnology, Jeonju University, Jeonju 55069, Korea
*Corresponding author: Younghoon Kim, Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea. Tel: +82-2-880-4808, E-mail: ykeys2584@snu.ac.kr

# These authors contributed equally to this work.

© Copyright 2025 Korean Society of Animal Science and Technology. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Aug 01, 2024; Revised: Sep 03, 2024; Accepted: Sep 03, 2024

Published Online: Jan 31, 2025

Abstract

MicroRNAs (miRNAs) are small noncoding RNAs that play a pivotal role in the regulation of gene expression. Analysis of miRNAs is important for understanding a variety of biological processes. Sequencing of miRNAs within milk-derived extracellular vesicles (EVs) provides valuable insights into the molecular mechanisms through which these EVs influence recipient cells. Comparative miRNA sequencing of colostrum and mature milk from different cow breeds can demonstrate breed-specific differences and improve the understanding of potential therapeutic applications in immune regulation and gut health. Therefore, this study was conducted to compare the miRNA profiles and characteristics of colostrum- and mature milk–derived EVs from Holstein and Jersey breeds and determine their effects on intestinal epithelial cells. The miRNA profiles of EVs isolated from the colostrum and mature milk of Holstein and Jersey cows were analyzed via small RNA sequencing. Holstein colostrum–derived EVs exhibited the most diverse miRNA profile with 421 identified miRNAs compared with 259 in mature milk–derived EVs. Jersey colostrum EVs had 198 miRNAs, whereas mature milk EVs had 282. Differential expression analysis revealed considerable miRNA differences between colostrum and mature milk, particularly in Holstein cows. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses revealed that miRNAs from colostrum EVs predominantly regulated immune-related pathways. Transcriptomic analysis of human colon cell line HT-29 treated with Holstein colostrum EVs confirmed the modulation of genes associated with immune responses. These findings indicate that colostrum-derived EVs, particularly from Holstein cows, play a pivotal role in immune regulation and could be potential candidates for therapeutic applications.

Keywords: MicroRNAs; Extracellular vesicles; Holstein; Jersey; Colostrum; Mature milk

INTRODUCTION

Extracellular vesicles (EVs) are nanovesicles that cells secrete into the extracellular environment and are characterized by their bilayer lipid membrane structure. Of these vesicles, the most well-known are exosomes, which are particles typically measuring 30–150 nm [1]. EVs, including exosomes, are found in various body fluids, such as blood, milk, cerebrospinal fluid, and saliva. They play a pivotal role in cell–cell interactions through the nucleic acids, proteins, metabolites, and lipids they contain [2]. These various bioactive substances in the vesicles are transmitted to the recipient cell, and this communication of information affects physiological processes, such as the regulation of immune responses, cell survival or death, and cell development [3]. Because of their potential for physiological regulation, EVs have recently been extensively studied in a range of fields.

One of the main components of EVs are microRNAs (miRNAs), which are small, noncoding RNAs. They are short, about 22 nucleotides (nt) in length, and have been found to play a pivotal role in regulating gene expression, leading to a growing body of research [4]. The complete set of miRNAs in a given cell or biological sample, known as the miRNome, is crucial for understanding the regulatory landscape within EVs [5]. Transported via EVs, miRNAs are involved in intercellular communication and immune regulation [6]. Specifically, miRNAs bind to target mRNAs and lead to their translation inhibition or degradation [7]. A milk-derived miRNA, miR-22-3p, has been reported to promote proliferation of human-derived intestinal epithelial cells [8]. Moreover, miR-31-5p enhanced endothelial cell proliferation and angiogenesis as well as alleviated diabetic wounds [9]. Understanding the miRNA profiles in EVs can provide insights into their roles in disease mechanisms, immune responses, and developmental processes.

Previous studies have reported that milk-derived EVs could affect intestinal epithelial cells. Exposure of milk EVs to HT-29 cells confirmed the uptake of milk EVs by these cells, accompanied by an elevation in miRNA levels within the EVs [10]. Exosomes derived from mouse breast milk promoted viability and proliferation of mouse intestinal epithelial cells [11]. Furthermore, EVs derived from human milk were reported to promote ZO-1 expression in Caco-2 cells and suppress the expression of inflammation-related genes [12]. These findings highlight the therapeutic potential of milk-derived EVs to promote gut health and modulate immune responses as well as the critical need for further comparative studies on EVs from different breeds or lactation stages to fully understand their biological functions and applications.

Colostrum, the milk secreted shortly after delivery, is important for the immunity and development of newborn animals, and its properties have been shown to be distinct from those of mature milk, and so are the properties of EVs. A comparison of EVs from colostrum, first milk, and mature milk showed that colostrum and first milk have more EVs than mature milk [13]. Differentially expressed milk-derived EVs across different phases and revealed the differences between human milk and bovine milk [14]. Colostrum- and mature milk–derived EVs have been shown to be able to protect intestinal epithelial cells from lipopolysaccharide (LPS)-induced damage and inflammation. Specifically, EVs from colostrum and mature milk have been found to exert differential effects on the expression of apoptosis-related and proinflammatory genes as well as to enhance cell proliferation and intestinal barrier function [15].

Holstein is the main breed for milk consumption, whereas Jersey is a recently introduced breed in Korea and is known for producing high-quality dairy products [16]. Although there have been several studies comparing the composition of milk between the two breeds, no comparative analysis of the characteristics of milk-derived EVs and miRNAs has been conducted. Therefore, this study aimed to compare the miRNAs and characteristics of colostrum- and mature milk–derived EVs from Holstein and Jersey breeds and to determine how EVs affect intestinal epithelial cells via transcriptomic analysis.

MATERIALS AND METHODS

Milk samples

All colostrum and mature milk samples were collected from three Holstein and three Jersey cows within 3 days and 1 month after delivery, respectively. To ensure comparability and control for breed-specific differences, these cows were raised on the same facility and fed the same diet. The pooled samples from each breed were immediately frozen and stored at −80°C until further analysis.

EV isolation

EVs were isolated from each milk sample using the previously described method, with slight modifications [17]. Briefly, the milk samples were centrifuged at 1,500×g for 10 min to remove the fat. Subsequently, the supernatant was centrifuged at 16,000×g for 1 h to pellet the cell debris. To obtain the whey fraction, the supernatant was centrifuged for an additional 1 h at 50,000×g. The whey fraction was ultracentrifuged at 100,000×g, and then the resulting supernatant was centrifuged at 135,000×g for 90 min to pellet the EVs. The centrifugation step was repeated, and phosphate buffered saline (PBS) was used to wash the EVs to ensure purity. The EVs were resuspended in PBS and stored at −80°C until further analysis.

miRNA extraction and sequencing

From each EVs, miRNAs were extracted as indicated using a QIAzol reagent (Qiagen, Hilden, Germany) and the miRNeasy Mini Kit (Qiagen). The purity and integrity of the extracted miRNAs were measured using the Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA). Subsequent library generation and sequencing were performed at Macrogen (Seoul, Korea). The SMARTer smRNA-Seq Kit for Illumina was used for library construction and sequenced using the HiSeq 2500 System (Illumina, San Diego, CA, USA). The generated raw data were filtered based on quality. The reads were sequentially aligned to reference genome, miRBase version 21 [18] and noncoding RNA database, RNAcentral 10.0. This alignment allowed for the classification of known miRNAs as well as other types of RNA, including tRNA, snRNA, and snoRNA. Novel miRNA prediction was performed using miRDeep2 [19]. The read counts for each miRNA were extracted from mapped miRNAs to report the abundance of each miRNA. Differentially expressed miRNAs (DEM) were determined by comparing across conditions each miRNA using edgeR of the R package with thresholds of |log2 fold change > 1| and p-value < 0.05. Representative miRNA target–binding sites were predicted using the TargetScan database (version 7.0). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotations were analyzed using the DAVID online tool (https://david.ncifcrf.gov/).

Cell culture and EV treatment

HT-29 cells obtained from ATCC were grown in RPMI 1640 medium (HyClone, Logan, UT, USA) supplemented with 10% fetal bovine serum. Complete monolayers of HT-29 cells were washed three times with PBS. Then, Holstein colostrum–derived EVs (HC;100 μg/mL) were added to the medium and incubated for 6 h at 37°C in the presence of 5% CO2. After exposure, the cells were washed with PBS, adherent cells were detached using a cell scraper, and total RNA was extracted using the RNeasy Mini Kit (Qiagen). The quality of total RNA was measured using the Agilent 2100 Bioanalyzer. The extracted RNA was stored at −80°C until further analysis.

RNA sequencing

For RNA-seq, libraries were prepared using the TruSeq RNA Sample Prep Kit version 2 (Illumina) according to the manufacturer’s protocol, and paired-end sequencing was performed using the Illumina HiSeq 2000 instrument. Sequenced raw data were trimmed using Trimmomatic 0.38 to remove adapter sequences, bases with quality < 3 from the ends of the reads, and bases that did not meet window size = 4 and mean quality = 15 using the sliding window trim technique. The trimmed data were generated by removing reads shorter than min length = 36 bp, and subsequent analysis were conducted using high-quality reads that based on the established quality controls. The Hisat2 version 2.1.0 program was used to build an index of the reference genome and paired-end clean reads of the human genome reference (GRCh38) were read and compared. Uniquely mapped reads were quantified with Subread/featureCounts version 1.5.1 using ENSEMBL version 82 transcriptome definitions. The generated data were subjected to differential expression analysis between samples using the R package edgeR, with genes corresponding to the thresholds |log2 fold change > 1| and p-value < 0.05 defined as significantly differentially expressed genes (DEG). GO and KEGG pathway annotations were analyzed using the DAVID online tool.

Statistical analysis

The identified miRNA profiles were visualized using EVenn (http://www.ehbio.com/test/venn). Statistical analyses and visualization were performed using GraphPad Prism 9.0 (San Diego, CA, USA).

RESULTS

Comparison of the identified miRNAs

To determine the miRNA profile of EVs present in each raw milk sample, EVs were isolated from each sample via ultracentrifugation. The miRNAs were extracted from the isolated EVs, and small RNA sequencing was performed using the HiSeq platform of Illumina (Fig. 1). A total of 421 miRNAs were identified in HC, and 259 miRNAs were sequenced in Holstein mature milk–derived EVs (HM). A total of 233 miRNAs were common to HC and HM, with 188 miRNAs identified in colostrum only and 26 miRNAs identified in mature milk only. Meanwhile, 198 and 282 miRNAs were identified in Jersey colostrum–derived EVs (JC) and Jersey mature milk–derived EVs (JM), respectively. There were 182 miRNAs that were common to both Jersey colostrum and mature milk, with 16 and 100 miRNAs identified in JC and JM, respectively.

jast-67-1-193-g1
Fig. 1. Comparison of the identified miRNAs. Venn diagram showing the number of cross-sample interactions for miRNAs present in EVs isolated from each sample. (A) HC vs. HM. (B) JC vs. JM. (C) HC vs. JC. (D) HM vs. JM. (E) All samples. HC, Holstein colostrum–derived extracellular vesicles; HM, Holstein mature milk–derived extracellular vesicles; JC, Jersey colostrum–derived extracellular vesicles; JM, Jersey mature milk–derived extracellular vesicles.
Download Original Figure

Next, the differences in miRNA profiles between the species were compared. The comparison revealed that 187 miRNAs were commonly expressed in both samples whereas 234 and 11 miRNAs were unique to HC and JC, respectively. Similarly, comparison between HM and JM revealed 233 shared miRNAs, with exclusive counts of 26 in HM and 49 in JM. Finally, comparison of the miRNA profiles of all samples revealed that 174 miRNAs were common across all samples. Unique miRNAs detected numbered 8, 9, and 16 in HM, JC, and JM, respectively. Contrarily, 150 miRNAs were exclusively found in HC, indicating that Holstein colostrum possesses the most distinct and extensive miRNA profile among the samples.

Comparison of miRNA profiles

Identification of miRNAs with expression levels exceeding 1% in each sample showed that 17 miRNAs were evenly expressed across the HC samples, with the top 7 accounting for about 66% of the total (Fig. 2). Meanwhile, seven miRNAs in JC had expression levels greater than 1%, whereas the two most highly expressed miRNAs accounted for about 65%. In mature milk EVs, 14 miRNAs in HM and 12 miRNAs in JM had expression levels greater than 1%. Interestingly, the top 6 miRNAs were the same across all samples except HC, although their proportions differed. In addition, the top 7 miRNAs in HC and JC were identical, comprising 76% and 78% of the total miRNA expression, respectively.

jast-67-1-193-g2
Fig. 2. Comparison of miRNA profiles. miRNAs present in more than 1% of the isolated EVs in each sample. (A) miRNA profiles of HC. (B) miRNA profiles of JC. (C) miRNA profiles of HM. (D) miRNA profiles of JM. HC, Holstein colostrum–derived extracellular vesicles; JC, Jersey colostrum–derived extracellular vesicles; HM, Holstein mature milk–derived extracellular vesicles; JM, Jersey mature milk–derived extracellular vesicles.
Download Original Figure
Comparison of the differential expression of miRNAs

To further analyze the miRNA profiles among samples, the identified miRNAs were filtered and normalized. As a result, 570 of 793 mature miRNAs were excluded, and 223 mature miRNAs were used for statistical analysis (Fig. 3). In comparison group C1, 60 miRNAs were upregulated and 67 were downregulated in HC compared with HM. In comparison group C2, 39 miRNAs were upregulated and 48 were downregulated in JC compared with JM. Interestingly, 68 miRNAs were upregulated and 70 were downregulated in JC compared with HC (C3). Contrarily, the comparison between HM and JM showed 9 upregulated and 20 downregulated miRNAs (C4). This suggests that the DEM between colostrum and mature milk are more pronounced in Holstein cows than in Jersey cows and that the species-specific differences are more evident in colostrum compared with mature milk.

jast-67-1-193-g3
Fig. 3. Comparison of the differential expressions of miRNAs. Volcano plots of differentially expressed miRNAs between samples. (A) HM vs. HC. (B) JM vs. JC. (C) HC vs. JC. (D) HM vs. JM. Colored dots were selected with cutoff criteria of p < 0.05 and absolute |log 2 (fold change)| > 1. HM, Holstein mature milk–derived extracellular vesicles; HC, Holstein colostrum–derived extracellular vesicles; JM, Jersey mature milk–derived extracellular vesicles; JC, Jersey colostrum–derived extracellular vesicles.
Download Original Figure
Target prediction for differentially expressed miRNAs

Gene enrichment analysis was conducted to predict the biological activities associated with the DEM for the three remaining combinations, except for C4, the mature milk comparison, which exhibited a slight difference in expression. The target genes of the top 10 DEM in each sample were predicted by TargetScan, and enrichment analysis of the selected gene set was conducted using the DAVID tool (Table 1). Immune-related pathways were found to be regulated in most comparative combinations. However, in both Holstein and Jersey breeds, these pathways were more prominently regulated in colostrum than in mature milk. In addition, comparison between JC and HC showed that more immune-related pathways were regulated in HC. Moreover, KEGG pathway analysis revealed that pathways related to MAPK signaling pathway and FoxO signaling pathway were upregulated, and cancer-related pathways were found to be upregulated in C1 and C2 rather than in C3. The top 10 pathways associated with immune responses are shown in Tables 2, 3, 4, 5, 6, and 7.

Table 1. Top 10 differentially expressed miRNAs between the two samples in each comparison
Top 10 DEMs of C1 Top 10 DEMs of C2 Top 10 DEMs of C3
Up in HC Up in HM Up in JC Up in JM Up in JC Up in HC
bta-miR-15a bta-miR-29d-3p bta-miR-101 bta-miR-135a bta-miR-885 bta-miR-143
bta-miR-222 bta-miR-29b bta-miR-455-3p bta-miR-326 bta-miR-1306 bta-miR-2284y
bta-miR-221 bta-miR-3613a bta-miR-490 bta-miR-2885 bta-miR-23b-5p bta-miR-30e-5p
bta-miR-30a-5p bta-miR-885 bta-miR-296-3p bta-miR-335 bta-miR-3613a bta-miR-2284x
bta-miR-142-3p bta-miR-23b-5p bta-miR-7857 bta-miR-2284x bta-miR-664b bta-miR-181c
bta-miR-182 bta-miR-6119-3p bta-miR-223 bta-miR-2284y bta-miR-150 bta-miR-769
bta-miR-142-5p bta-miR-2904 bta-miR-130a bta-miR-6522 bta-miR-455-3p bta-miR-6522
bta-miR-3431 bta-miR-1306 bta-miR-671 bta-miR-30e-5p bta-miR-365-3p bta-miR-326
bta-miR-320b bta-miR-664b bta-miR-744 bta-miR-2313-3p bta-miR-1246 bta-miR-874
bta-miR-181a bta-miR-150 bta-miR-15a bta-miR-196b bta-miR-6119-3p bta-miR-2336

DEMs, differentially expressed miRNAs; C1, comparison between HC and HM; C2, comparison between JC and JM; C3, comparison between JC and HC; HC, Holstein colostrum–derived extracellular vesicles; HM, Holstein mature milk–derived extracellular vesicles; JC, Jersey colostrum–derived extracellular vesicles; JM, Jersey mature milk–derived extracellular vesicles.

Download Excel Table
Table 2. Gene Ontology and KEGG pathway enrichment analyses of the top 10 differentially expressed miRNAs of Holstein colostrum–derived extracellular vesicles in the comparison between Holstein colostrum– and Holstein mature milk–derived extracellular vesicles
GO ID GO term p-value Pathway ID Definition p-value
GO:0042981 Regulation of apoptotic process 0.003 bta04010 MAPK signaling pathway < 0.001
GO:0008285 Negative regulation of cell proliferation 0.004 bta04014 Ras signaling pathway < 0.001
GO:0001525 Angiogenesis 0.007 bta05224 Breast cancer 0.003
GO:0007267 Cell–cell signaling 0.008 bta04935 Growth hormone synthesis, secretion, and action 0.006
GO:0045930 Negative regulation of mitotic cell cycle 0.013 bta05200 Pathways in cancer 0.011
GO:0030334 Regulation of cell migration 0.014 bta05210 Colorectal cancer 0.013
GO:0000422 Mitophagy 0.020 bta04062 Chemokine signaling pathway 0.019
GO:0042771 Intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediator 0.020 bta05100 Bacterial invasion of epithelial cells 0.020
GO:0043524 Negative regulation of neuronal apoptotic process 0.021 bta04022 cGMP-PKG signaling pathway 0.022
GO:0000045 Autophagosome assembly 0.023 bta04211 Longevity regulating pathway 0.042

Only the top 10 GO terms representing the biological process and KEGG pathway are listed in the table.

KEGG, Kyoto Encyclopedia of Genes and Genomes; MAPK, mitogen-activated protein kinase; cGMP-PKG, cyclic guanosine monophosphate–protein kinase G.

Download Excel Table
Table 3. Gene Ontology and KEGG pathway enrichment analyses of the top 10 differentially expressed miRNAs of Holstein mature milk–derived extracellular vesicles in the comparison between Holstein colostrum– and Holstein mature milk–derived extracellular vesicles
GO ID GO term p-value Pathway ID Definition p-value
GO:0030198 Extracellular matrix organization < 0.001 bta04974 Protein digestion and absorption < 0.001
GO:0001836 Release of cytochrome c from the mitochondria 0.001 bta04510 Focal adhesion < 0.001
GO:0097192 Extrinsic apoptotic signaling pathway in the absence of ligand 0.003 bta04151 PI3K-Akt signaling pathway < 0.001
GO:0071456 Cellular response to hypoxia 0.025 bta04512 ECM–receptor interaction < 0.001
GO:0051781 Positive regulation of cell division 0.034 bta04926 Relaxin signaling pathway 0.001
GO:0045893 Positive regulation of transcription, DNA-templated 0.035 bta05165 Human papillomavirus infection 0.001
GO:0060070 Canonical Wnt signaling pathway 0.036 bta05222 Small cell lung cancer 0.001
GO:0051897 Positive regulation of protein kinase B signaling 0.037 bta05200 Pathways in cancer 0.005
GO:0010595 Positive regulation of endothelial cell migration 0.043 bta01521 EGFR tyrosine kinase inhibitor resistance 0.009
GO:0008630 Intrinsic apoptotic signaling pathway in response to DNA damage 0.049 bta05205 Proteoglycans in cancer 0.018

Only the top 10 GO terms representing the biological process and KEGG pathway are listed in the table.

KEGG, Kyoto Encyclopedia of Genes and Genomes; PI3K, phosphatidylinositol-3-kinase; ECM, extracellular matrix; EGFR, epidermal growth factor receptor.

Download Excel Table
Table 4. Gene Ontology and KEGG pathway enrichment analyses of the top 10 differentially expressed miRNAs of Jersey colostrum–derived extracellular vesicles in the comparison between Jersey colostrum– and Jersey mature milk–derived extracellular vesicles
GO ID GO term p-value Pathway ID Definition p-value
GO:0008285 Negative regulation of cell proliferation 0.001 bta04150 mTOR signaling pathway < 0.001
GO:0016310 Phosphorylation 0.002 bta04010 MAPK signaling pathway 0.001
GO:0045669 Positive regulation of osteoblast differentiation 0.006 bta04068 FoxO signaling pathway 0.001
GO:0030178 Negative regulation of the Wnt signaling pathway 0.006 bta05224 Breast cancer 0.003
GO:0001764 Neuronal migration 0.008 bta05202 Transcriptional misregulation in cancer 0.007
GO:0006915 Apoptotic process 0.010 bta05225 Hepatocellular carcinoma 0.008
GO:0001935 Endothelial cell proliferation 0.012 bta05210 Colorectal cancer 0.010
GO:0051726 Regulation of cell cycle 0.017 bta05223 Nonsmall cell lung cancer 0.014
GO:0030154 Cell differentiation 0.018 bta05200 Pathways in cancer 0.022
GO:0035556 Intracellular signal transduction 0.023 bta04950 Maturity onset diabetes of the young 0.033

Only the top 10 GO terms representing the biological process and KEGG pathway are listed in the table.

KEGG, Kyoto Encyclopedia of Genes and Genomes; mTOR, mammalian target of rapamycin; MAPK, mitogen-activated protein kinase; FoxO, forkhead box O.

Download Excel Table
Table 5. Gene Ontology and KEGG pathway enrichment analyses of the top 10 differentially expressed miRNAs of Jersey mature milk–derived extracellular vesicles in the comparison between Jersey colostrum– and Jersey mature milk–derived extracellular vesicles
GO ID GO term p-value Pathway ID Definition p-value
GO:0034599 Cellular response to oxidative stress < 0.001 bta04010 MAPK signaling pathway 0.001
GO:0043162 Ubiquitin-dependent protein catabolic process via the multivesicular body sorting pathway 0.002 bta05235 PD-L1 expression and PD-1 checkpoint pathway in cancer 0.002
GO:0071333 Cellular response to glucose stimulus 0.002 bta04360 Axon guidance 0.003
GO:0010458 Exit from mitosis 0.002 bta04912 GnRH signaling pathway 0.006
GO:0010508 Positive regulation of autophagy 0.003 bta04218 Cellular senescence 0.009
GO:0071805 Potassium ion transmembrane transport 0.004 bta04921 Oxytocin signaling pathway 0.017
GO:0010737 Protein kinase A signaling 0.005 bta04068 FoxO signaling pathway 0.017
GO:0009408 Response to heat 0.011 bta04720 Long-term potentiation 0.017
GO:0010766 Negative regulation of sodium ion transport 0.011 bta04550 Signaling pathways regulating pluripotency of stem cells 0.028
GO:0030036 Actin cytoskeleton organization 0.017 bta05216 Thyroid cancer 0.031

Only the top 10 GO terms representing the biological process and KEGG pathway are listed in the table.

KEGG, Kyoto Encyclopedia of Genes and Genomes; MAPK, mitogen-activated protein kinase; PD-L1, programmed death ligand 1; PD-1, programmed cell death protein 1; GnRH, gonadotropin-releasing hormone; FoxO, forkhead box O.

Download Excel Table
Table 6. Gene Ontology and KEGG pathway enrichment analyses of the top 10 differentially expressed miRNAs of Jersey colostrum–derived extracellular vesicles in the comparison between Jersey colostrum– and Holstein colostrum–derived extracellular vesicles
GO ID GO term p-value Pathway ID Definition p-value
GO:0007218 Neuropeptide signaling pathway 0.007 bta04550 Signaling pathways regulating the pluripotency of stem cells 0.005
GO:0051897 Positive regulation of protein kinase B signaling 0.007
GO:0060216 Definitive hemopoiesis 0.014
GO:0043922 Negative regulation by host of viral transcription 0.018
GO:0045165 Cell fate commitment 0.038
GO:0046326 Positive regulation of glucose import 0.043
GO:0042981 Regulation of apoptotic process 0.044
GO:0070374 Positive regulation of ERK1 and ERK2 cascade 0.044
GO:0048856 Anatomical structure development 0.044
GO:0060129 Thyroid-stimulating hormone-secreting cell differentiation 0.045

Only the top 10 GO terms representing the biological process and KEGG pathway are listed in the table.

KEGG, Kyoto Encyclopedia of Genes and Genomes; ERK, extracellular signal–regulated kinase.

Download Excel Table
Table 7. Gene Ontology and KEGG pathway enrichment analyses of the top 10 differentially expressed miRNAs of Holstein colostrum–derived extracellular vesicles in the comparison between Jersey colostrum– and Holstein colostrum–derived extracellular vesicles
GO ID GO term p-value Pathway ID Definition p-value
GO:0071805 Potassium ion transmembrane transport 0.004 bta04371 Apelin signaling pathway 0.008
GO:0010633 Negative regulation of epithelial cell migration 0.007 bta04211 Longevity regulating pathway 0.016
GO:0030036 Actin cytoskeleton organization 0.008 bta04151 PI3K-Akt signaling pathway 0.024
GO:0001558 Regulation of cell growth 0.008 bta00330 Arginine and proline metabolism 0.046
GO:0007346 Regulation of mitotic cell cycle 0.016
GO:0030837 Negative regulation of actin filament polymerization 0.018
GO:0071542 Dopaminergic neuron differentiation 0.021
GO:0006936 Muscle contraction 0.022
GO:0010508 Positive regulation of autophagy 0.024
GO:0042752 Regulation of circadian rhythm 0.024

Only the top 10 GO terms representing the biological process and KEGG pathway are listed in the table.

KEGG, Kyoto Encyclopedia of Genes and Genomes; PI3K, phosphatidylinositol-3-kinase.

Download Excel Table
Target prediction for the top 7 miRNAs of HC

As HC had the most characteristic miRNA profile, GO and KEGG enrichment analyses were conducted on the top 7 miRNAs that accounted for more than 3% of abundance. Among the enrichment results, a total of 37 items related to biological processes, 15 related to cellular components, and 15 related to molecular functions were identified as important. KEGG pathway analysis revealed that a total of 31 pathways were significantly matched, most of which were related to immunity or cancer. The top 10 pathways associated with immune responses are shown in Table 8.

Table 8. Gene Ontology and KEGG pathway enrichment analyses of the top 7 miRNAs of Holstein colostrum–derived extracellular vesicles
GO ID GO term p-value Pathway ID Definition p-value
GO:0001764 Neuronal migration < 0.001 bta05200 Pathways in cancer < 0.001
GO:0006468 Protein phosphorylation < 0.001 bta04068 FoxO signaling pathway < 0.001
GO:0010718 Positive regulation of epithelial to mesenchymal transition 0.002 bta04350 TGF-beta signaling pathway < 0.001
GO:0015031 Protein transport 0.002 bta05224 Breast cancer < 0.001
GO:0043491 Protein kinase B signaling 0.003 bta04010 MAPK signaling pathway 0.001
GO:0008285 Negative regulation of cell proliferation 0.004 bta05210 Colorectal cancer 0.001
GO:0030177 Positive regulation of the Wnt signaling pathway 0.009 bta05225 Hepatocellular carcinoma 0.004
GO:0043065 Positive regulation of apoptotic process 0.014 bta05226 Gastric cancer 0.007
GO:0035556 Intracellular signal transduction 0.017 bta04510 Focal adhesion 0.012
GO:0098609 Cell–cell adhesion 0.037 bta04014 Ras signaling pathway 0.014

Only the top 10 GO terms representing the biological process and KEGG pathway are listed in the table.

KEGG, Kyoto Encyclopedia of Genes and Genomes; FoxO, forkhead box O; TGF, transforming growth factor; MAPK, mitogen-activated protein kinase.

Download Excel Table
Transcriptome analysis of EV-treated HT-29 cells

Based on the previous results, HC is the most likely to regulate immune-related functions in the sample. Therefore, to confirm the effect of HC, human colon cell line HT-29 was treated with HC and subjected to transcriptomic analysis. DEG analysis revealed that of a total of 2,892 genes, 203 and 255 were upregulated and downregulated, respectively, upon treatment with HC. Gene enrichment analysis of DEG was conducted, and the results are shown in Table 9. GO analysis revealed that the upregulated genes were related to chemotaxis and negative regulation of the macrophage cytokine production pathway. Downregulated genes were identified as specific genes for negative regulation of platelet aggregation, telomere maintenance in response to DNA damage, regulation of cell cycle G2/M phase transition pathway. KEGG pathway analysis revealed that the upregulated genes did not produce substantial results and that the downregulated genes were associated with the NF-kappa B signaling and metabolic pathways.

Table 9. Gene Ontology and KEGG pathway enrichment analyses of differentially expressed genes between negative control and Holstein colostrum–derived extracellular vesicles
Upregulated Downregulated
ID Definition p-value ID Definition p-value
GO_BP:0006935 Chemotaxis 0.002 GO_BP:0090331 Negative regulation of platelet aggregation 0.002
GO_BP:0010936 Negative regulation of macrophage cytokine production 0.048 GO_BP:0090258 Negative regulation of mitochondrial fission 0.022
GO_CC:0005886 Plasma membrane 0.010 GO_BP:0043247 Telomere maintenance in response to DNA damage 0.038
GO_CC:0005615 Extracellular space 0.022 GO_BP:1902749 Regulation of cell cycle G2/M phase transition 0.038
GO_MF:0016787 Hydrolase activity 0.005
GO_MF:0004672 Protein kinase activity 0.020
KEGG: hsa04064 NF-kappa B signaling pathway 0.017
KEGG: hsa01100 Metabolic pathways 0.046

KEGG, Kyoto Encyclopedia of Genes and Genomes; BP, biological process; CC, cellular component; MF, molecular function; NF, nuclear factor.

Download Excel Table

DISCUSSION

In this study, the miRNA characteristics of EVs derived from colostrum and mature milk of two cattle breeds were compared. The comparison revealed that the most diverse miRNAs were evenly present in Holstein colostrum EVs whereas fewer miRNAs were identified in Jersey colostrum. For the miRNA profiles of mature milk samples, the top 7 miRNAs were found to be identical in Holstein and Jersey, accounting for approximately 77% of the miRNAs. The DEM analysis revealed more miRNA differences in the comparison between mature milk and colostrum, consistent with the miRNA profile results, and few DEMs were found in the comparison between mature milk miRNAs. Gene enrichment analysis of the target genes of the top 10 DEM revealed that immune-related pathways were involved in colostrum than in mature milk in both Holstein and Jersey breeds. Holstein colostrum EVs with the most distinctive miRNA signatures were further analyzed by transcriptomics for their effects on intestinal epithelial cells, and genes associated with immune-related pathways were found to be modulated. These results indicate that EVs derived from Holstein and Jersey colostrum, rather than mature milk, are enriched in miRNAs that regulate immune-related pathways and that Holstein colostrum is the most likely immune modulator among them.

miRNAs are known to play a pivotal role in biological processes by regulating gene expression. Due to this property, research on the importance of miRNAs is rapidly growing [2022]. Previous studies have demonstrated that miRNAs in bovine can influence gene expression in humans [10] and are also used to ameliorate diseases or for the diagnosis of diseases as biomarkers [2325]. The bioactive functions of EVs are closely associated with their internal miRNAs [26]. Therefore, the identification of the profile of miRNAs present in EVs is important for EV characterization.

Most dairy cows currently bred are Holstein, which are characterized by their large body size and higher milk yield compared with other breeds [27]. Jersey cows are characterized by their smaller body size and lower feed consumption compared with Holstein [28]. They are also more heat-resistant than Holstein cows and have a higher amount of protein and fat in their milk, which makes them more efficient in the production of dairy products [16]. Most farmers breed Holstein for efficient milk production, and thus, most of the milk consumed by humans comes from Holstein, but Jersey cows are increasingly being domesticated to produce high-quality dairy products [29]. However, most comparisons of Holstein and Jersey milk have focused on their nutritional composition, and the miRNA content has not been investigated. To the best of our knowledge, this study is the first to explore the profiles of EV-derived miRNAs in colostrum and mature milk between Jersey and Holstein cows. The miRNA profiles revealed that in Holstein breeds, more miRNAs were identified in colostrum-derived EVs than in mature milk EVs. Meanwhile, in Jersey milk, more miRNAs were detected in mature milk–derived EVs than in colostrum-derived ones. In a previous study, miRNA analysis of colostrum and mature milk from Holstein and Doğu Anadolu Kirmizisi cows revealed that in both breeds, known miRNAs were more highly expressed in mature milk than in colostrum [30]. Other studies have investigated miRNAs in bovine and porcine breast milk and detected about 100 more miRNAs in colostrum than in mature milk [31,32].

Jersey milk is known to have higher protein and lipid contents than Holstein milk. Previous studies have demonstrated that milk with higher ratios of fat and protein is enriched in miRNAs that regulate protein and lipid metabolism, mammary gland development, and amino acid biosynthesis [33,34]. However, the present study found fewer differences in miRNAs in the mature milk samples and no significant association with protein and fat metabolism compared with the colostrum samples. A previous study reported that colostrum from Holstein and Jersey breeds did not differ in terms of fat and protein contents [29]. To confirm the association between miRNA analysis results and milk composition, further compositional analysis of milk samples is warranted. Moreover, the results are controversial because the miRNA profiles of colostrum and mature milk have not been sufficiently studied yet, and the expression level of miRNAs may be influenced by the state of the individual from which they are derived, sample management, and time and duration of lactation [35]. To more accurately compare the miRNA profiles, it is important to use a wide range of samples under various conditions.

Previous studies have demonstrated that miRNAs in colostrum are more involved in immunologic pathways than miRNAs in mature milk [36,37]. Analysis of miRNAs in porcine breast milk has revealed that there are more immune-related miRNAs in colostrum and that these miRNAs are present in the serum of piglets [32]. The results of the present study indicated that Holstein colostrum compared with mature milk was associated with biological activities, such as regulation of apoptotic process, negative regulation of cell proliferation, and cell–cell signaling, as well as KEGG pathways, such as MAPK, Ras, and chemokine signaling pathways. Similarly, the target genes of Jersey colostrum miRNAs compared with mature milk were predicted to be associated with biological processes, such as negative regulation of cell proliferation, phosphorylation, negative regulation of Wnt signaling pathway, and KEGG pathways, such as mTOR signaling pathway. Meanwhile, the target pathways of DEMs in HC and JC were predicted to regulate more immune-related processes in HC than in JC. In addition, the KEGG pathway analysis revealed that both upregulated and downregulated miRNAs in each comparison combination would affect different types of cancer and immune-related pathways. These results are consistent with previous studies demonstrating that milk miRNAs are strongly associated with cancer and immunity [22].

The most highly expressed miRNAs in Holstein colostrum include bta-miR-26a, bta-miR-30a-5p, bta-miR-181a, and bta-let-7a-5p. miR-26-a regulates the secretion of proinflammatory cytokines in LPS-stimulated macrophages [38]. Moreover, miR-30a-5p and miR-26a have been reported to be capable of alleviating oncogenic inflammation in the colon by regulating the expressions of NF-κB and IL-6 [39,40]. miR-181a has an anti-inflammatory activity mediated by the inhibition of IL1a in LPS-treated monocytes and macrophages [41]. In addition, let-7a-5p regulates IL-10 secretion in the inflammatory environment; it improved lung function by reducing collagen deposition and macrophage infiltration in mice with acute lung injury [42]. Together with the previous findings, further studies are warranted to confirm the possible anti-inflammatory functions of miRNAs with high proportions in HC and the overall role of HC. Furthermore, miRNAs typically do not bind directly to DNA, but instead inhibit gene expression by attaching to complementary sequences of the target mRNA and inhibiting translation or degrading the mRNA itself. This mode of action carries a low risk of causing direct mutations in DNA sequences [6]. However, the long-term effects of miRNA therapy and whether it may have unintended consequences are still unclear and need further study and long-term observation.

EVs are characterized by their ability to interact with other cells through their internal miRNAs and other bioactive substances. Previous studies have demonstrated that EVs are taken up by target cells by pathways such as phagocytosis, endocytosis, and membrane fusion [43]. Milk-derived EVs have been shown to be taken up by Caco-2 cells in a concentration-dependent manner and are not cytotoxic even after 6 h of EV treatment [44]. It has also been confirmed that milk EVs are taken up by differentiated human macrophages [45]. In this study, the effects of HC on the human colon cancer cell line HT-29 were investigated using a transcriptomics approach. It was found that most of the regulated pathways were associated with immunity, with more downregulation than upregulation. However, there were not many significantly regulated pathways and matched genes, which may be due to the concentration and time of exposure of EVs [10].

CONCLUSION

In conclusion, this study identified differences in the miRNA profiles of HC, HM, JC, and JM, with the latter showing less significant differences between species, whereas more diverse miRNAs were characterized in Holstein than Jersey colostrum EVs. Interestingly, regardless of the species, more immune-related pathways were predicted to be regulated in colostrum EVs compared with mature milk EVs. Notably, Holstein colostrum had the most distinctive miRNA profile, and it was shown that immune-related metabolic pathways were regulated when exposed to human intestinal epithelial cells. It is suggested that analysis of the gene–gene interactions through miRNA profiling and target gene analysis is the cornerstone for screening candidate miRNAs for disease amelioration and identifying the potential of EVs.

Competing interests

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

Funding sources

This research was supported by a National Research Foundation of Korea Grant, funded by the Korean government (MEST) (NRF-2021R1A2C3011051), by the Korea Evaluation Institute of Industrial Technology (KEIT, 20012411), and by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2022M3A9I5018286).

Acknowledgements

Not applicable.

Availability of data and material

Upon reasonable request, the datasets of this study can be available from the corresponding author.

Authors’ contributions

Conceptualization: Mun D, Ryu S, Lim DH, Oh S, Kim Y.

Data curation: Mun D, Ryu S, Lim DH, Oh S, Kim Y.

Formal analysis: Mun D, Ryu S.

Methodology: Mun D, Ryu S, Lim DH, Kim Y.

Software: Mun D, Ryu S.

Validation: Mun D, Ryu S, Lim DH, Oh S, Kim Y.

Investigation: Mun D, Ryu S, Lim DH, Oh S, Kim Y.

Writing-original draft: Mun D, Ryu S, Lim DH, Oh S, Kim Y.

Writing-review & editing: Mun D, Ryu S, Lim DH, Oh S, Kim Y.

Ethics approval and consent to participate

This article does not require IRB/IACUC approval because there are no human and animal participants.

REFERENCES

1.

Tkach M, Théry C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016; 164:1226-32

2.

Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014; 14:195-208

3.

Yáñez-Mó M, Siljander PRM, Andreu Z, Zavec AB, Borràs FE, Buzas EI, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015; 4:27066

4.

Munir J, Lee M, Ryu S. Exosomes in food: health benefits and clinical relevance in diseases. Adv Nutr. 2020; 11:687-96

5.

Pereira A, Moreira F, Vinasco-Sandoval T, Cunha A, Vidal A, Ribeiro-dos-Santos AM, et al. miRNome reveals new insights into the molecular biology of field cancerization in gastric cancer. Front Genet. 2019; 10:592

6.

Ishida M, Selaru FM. miRNA-based therapeutic strategies. Curr Pathobiol Rep. 2013; 1:63-70

7.

Soifer HS, Rossi JJ, Saetrom P. MicroRNAs in disease and potential therapeutic applications. Mol Ther. 2007; 15:2070-9

8.

Jiang R, Lönnerdal B. Milk-derived miR-22-3p promotes proliferation of human intestinal epithelial cells (HIECs) by regulating gene expression. Nutrients. 2022; 14:4901

9.

Yan C, Chen J, Wang C, Yuan M, Kang Y, Wu Z, et al. Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Deliv. 2022; 29:214-28

10.

López de Las Hazas MC, Del Pozo-Acebo L, Hansen MS, Gil-Zamorano J, Mantilla-Escalante DC, Gómez-Coronado D, et al. Dietary bovine milk miRNAs transported in extracellular vesicles are partially stable during GI digestion, are bioavailable and reach target tissues but need a minimum dose to impact on gene expression. Eur J Nutr. 2022; 61:1043-56

11.

Hock A, Miyake H, Li B, Lee C, Ermini L, Koike Y, et al. Breast milk-derived exosomes promote intestinal epithelial cell growth. J Pediatr Surg. 2017; 52:755-9

12.

Chiba T, Maeda T. Human milk exosomes induce ZO-1 expression via inhibition of REDD1 expression in human intestinal epithelial cells. Biol Pharm Bull. 2023; 46:893-7

13.

Santoro J, Mukhopadhya A, Oliver C, Brodkorb A, Giblin L, O’Driscoll L. An investigation of extracellular vesicles in bovine colostrum, first milk and milk over the lactation curve. Food Chem. 2023; 401:134029

14.

Yang M, Song D, Cao X, Wu R, Liu B, Ye W, et al. Comparative proteomic analysis of milk-derived exosomes in human and bovine colostrum and mature milk samples by iTRAQ-coupled LC-MS/MS. Food Res Int. 2017; 92:17-25

15.

Liu W, Du C, Nan L, Li C, Wang H, Fan Y, et al. The difference of milk-derived extracellular vesicles from cow colostrum and mature milk on miRNAs expression and protecting intestinal epithelial cells against lipopolysaccharide damage. Int J Mol Sci. 2024; 25:3880

16.

Yoo J, Song M, Park W, Oh S, Ham JS, Jeong SG, et al. A comparison of quality characteristics in dairy products made from Jersey and Holstein milk. Food Sci Anim Resour. 2019; 39:255-65

17.

Maburutse BE, Park MR, Oh S, Kim Y. Evaluation and characterization of milk-derived microvescicle isolated from bovine colostrum. Korean J Food Sci Anim. 2017; 37:654-62

18.

Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006; 34:D140-4

19.

Friedländer MR, Mackowiak SD, Li N, Chen W, Rajewsky N. miRDeep2 accurately identifies known and hundreds of novel microRNA genes in seven animal clades. Nucleic Acids Res. 2012; 40:37-52

20.

Melnik BC, Kakulas F, Geddes DT, Hartmann PE, John SM, Carrera-Bastos P, et al. Milk miRNAs: simple nutrients or systemic functional regulators?. Nutr Metab (Lond). 2016; 13:42

21.

Golan-Gerstl R, Elbaum Shiff Y, Moshayoff V, Schecter D, Leshkowitz D, Reif S. Characterization and biological function of milk-derived miRNAs. Mol Nutr Food Res. 2017; 61:1700009

22.

Benmoussa A, Provost P. Milk microRNAs in health and disease. Compr Rev Food Sci Food Saf. 2019; 18:703-22

23.

Kumarswamy R, Volkmann I, Thum T. Regulation and function of miRNA-21 in health and disease. RNA Biol. 2011; 8:706-13

24.

Backes C, Meese E, Keller A. Specific miRNA disease biomarkers in blood, serum and plasma: challenges and prospects. Mol Diagn Ther. 2016; 20:509-18

25.

Iannaccone M, Cosenza G, Pauciullo A, Garofalo F, Proroga YT, Capuano F, et al. Milk microRNA-146a as a potential biomarker in bovine tuberculosis. J Dairy Res. 2018; 85:178-80

26.

He S, Huang L, Shao C, Nie T, Xia L, Cui B, et al. Several miRNAs derived from serum extracellular vesicles are potential biomarkers for early diagnosis and progression of Parkinson’s disease. Transl Neurodegener. 2021; 10:25

27.

Le Guillou S, Leduc A, Laubier J, Barbey S, Rossignol MN, Lefebvre R, et al. Characterization of Holstein and Normande whole milk miRNomes highlights breed specificities. Sci Rep. 2019; 9:20345

28.

Lim DH, Mayakrishnan V, Ki KS, Kim Y, Kim TI. The effect of seasonal thermal stress on milk production and milk compositions of Korean Holstein and Jersey cows. Anim Biosci. 2021; 34:567-74

29.

Lim DH, Mayakrishnan V, Lee HJ, Ki KS, Kim TI, Kim Y. A comparative study on milk composition of Jersey and Holstein dairy cows during the early lactation. J Anim Sci Technol. 2020; 62:565-76

30.

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

31.

Chen X, Gao C, Li H, Huang L, Sun Q, Dong Y, et al. Identification and characterization of microRNAs in raw milk during different periods of lactation, commercial fluid, and powdered milk products. Cell Res. 2010; 20:1128-37

32.

Gu Y, Li M, Wang T, Liang Y, Zhong Z, Wang X, et al. Lactation-related microRNA expression profiles of porcine breast milk exosomes. PLOS ONE. 2012; 7e43691

33.

Wang X, Zhang L, Jin J, Xia A, Wang C, Cui Y, et al. Comparative transcriptome analysis to investigate the potential role of miRNAs in milk protein/fat quality. Sci Rep. 2018; 8:6250

34.

Cui X, Zhang S, Zhang Q, Guo X, Wu C, Yao M, et al. Comprehensive microRNA expression profile of the mammary gland in lactating dairy cows with extremely different milk protein and fat percentages. Front Genet. 2020; 11:548268

35.

Izumi H, Kosaka N, Shimizu T, Sekine K, Ochiya T, Takase M. Time-dependent expression profiles of microRNAs and mRNAs in rat milk whey. PLOS ONE. 2014; 9e88843

36.

Izumi H, Kosaka N, Shimizu T, Sekine K, Ochiya T, Takase M. Bovine milk contains microRNA and messenger RNA that are stable under degradative conditions. J Dairy Sci. 2012; 95:4831-41

37.

Zhou Q, Li M, Wang X, Li Q, Wang T, Zhu Q, et al. Immune-related microRNAs are abundant in breast milk exosomes. Int J Biol Sci. 2012; 8:118-23

38.

Yu T, Wang P, Wu Y, Zhong J, Chen Q, Wang D, et al. MiR-26a reduces inflammatory responses via inhibition of PGE2 production by targeting COX-2. Inflammation. 2022; 45:1484-95

39.

Zhang W, Fu X, Xie J, Pan H, Han W, Huang W. miR-26a attenuates colitis and colitis-associated cancer by targeting the multiple intestinal inflammatory pathways. Mol Ther Nucleic Acids. 2021; 24:264-73

40.

Zhang C, Zheng J, Han X, Zhao J, Cheng S, Li A. Bovine colostrum miR-30a-5p targets the NF-κB signaling pathway to alleviate inflammation in intestinal epithelial cells. J Agric Food Chem. 2024; 72:9164-78

41.

Xie W, Li M, Xu N, Lv Q, Huang N, He J, et al. miR-181a regulates inflammation responses in monocytes and macrophages. PLOS ONE. 2013; 8e58639

42.

Chen SY, Chen YL, Li PC, Cheng TS, Chu YS, Shen YS, et al. Engineered extracellular vesicles carrying let-7a-5p for alleviating inflammation in acute lung injury. J Biomed Sci. 2024; 31:30

43.

Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014; 3:24641

44.

Hansen MS, Gadegaard ISE, Arnspang EC, Blans K, Nejsum LN, Rasmussen JT. Specific and non-invasive fluorescent labelling of extracellular vesicles for evaluation of intracellular processing by intestinal epithelial cells. Biomedicines. 2020; 8:211

45.

Izumi H, Tsuda M, Sato Y, Kosaka N, Ochiya T, Iwamoto H, et al. Bovine milk exosomes contain microRNA and mRNA and are taken up by human macrophages. J Dairy Sci. 2015; 98:2920-33