Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery
Abstract
:1. Introduction
1.1. Extracellular Vesicles
1.2. EV Cargo
1.3. EVs in Biofluids
2. Milk as a Biofluid in Interindividual and Cross-Species Communication
2.1. Milk EVs Mediate Intercellular Communication
2.2. Milk EV Cargo
3. Milk EVs in Cross-Organism and Cross-Species Communication
3.1. Milk EVs Mediate Post-Natal Development and Growth
3.2. Milk EVs Have Immunoregulatory Effect
3.3. Milk EVs Have Role in Physiological Processes, Health and Disease
4. Milk EVs in Therapy
4.1. Milk EVs as Drug-Delivery Systems
4.2. Milk EVs as Anti-Inflammatory Agents
5. Future Perspective
Author Contributions
Acknowledgments
Conflicts of Interest
Abbreviations
AKT | Protein kinase B |
CD | Cluster of differentiation |
DC-SIGN | Dendritic cell-specific intercellular adhesion molecule-grabbing non-integrin |
DNMT | DNA methyltransferase |
EGFR | Epidermal growth factor receptor |
EMT | Epithelial-mesenchymal transition |
EVs | Extracellular vesicles |
FABP4 | Fatty acid binding protein 4 |
FGF-2 | Fibroblast growth factor 2 |
FoxP3 | Forkhead box P3 |
FTO | Fat-mass and obesity associated protein |
GM3 | monosialodihexosylganglioside |
HSP | Heat-shock protein |
IECs | Intestinal epithelial cells |
IFN-γ | Interferon gamma |
IGF-1 | Insulin-like growth factor-1 |
IL | Interleukin |
INS | Insulin |
MFG | Milk fat globule |
MHC | Major histocompatibility complex |
miR/miRNA | Micro RNA |
mTORC1 | Mammalian target of rapamycin complex 1 |
MUC-1 | Mucin-1 |
MVB | Multivesicular body |
PBMC | Peripheral blood mononuclear cell |
ROCK1 | Rho-associated, coiled-coiled containing protein kinase-1 |
Runx2 | Runt related transcription factor 2 |
SMAD2/3 | Mothers against decapentaplegic homolog 2/3 |
SREBP1 | Sterol regulatory element-binding protein 1 |
TGF-β | Transforming growth factor-β |
TLR | Toll-like receptor |
TNF-α | Tumor necrosis factor-α |
Tregs | Regulatory T cells |
TSG101 | Tumor susceptibility gene 101 |
WISP-1 | Wnt1-inducible-signaling pathway protein-1 |
References
- Kalra, H.; Drummen, G.; Mathivanan, S. Focus on Extracellular Vesicles: Introducing the Next Small Big Thing. Int. J. Mol. Sci. 2016, 17, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Boil. 2018, 19, 213–228. [Google Scholar] [CrossRef] [PubMed]
- Hristov, M.; Erl, W.; Linder, S.; Weber, P.C. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood 2004, 104, 2761–2766. [Google Scholar] [CrossRef] [PubMed]
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Boil. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
- Harding, C.; Stahl, P. Transferrin recycling in reticulocytes: pH and iron are important determinants of ligand binding and processing. Biochem. Biophys. Res. Commun. 1983, 113, 650–658. [Google Scholar] [CrossRef]
- Pan, B.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef]
- Cocucci, E.; Racchetti, G.; Meldolesi, J. Shedding microvesicles: Artefacts no more. Trends Cell Boil. 2009, 19, 43–51. [Google Scholar] [CrossRef]
- Zarà, M.; Guidetti, G.F.; Camera, M.; Canobbio, I.; Amadio, P.; Torti, M.; Tremoli, E.; Barbieri, S.S. Biology and Role of Extracellular Vesicles (EVs) in the Pathogenesis of Thrombosis. Int. J. Mol. Sci. 2019, 20, 2840. [Google Scholar] [CrossRef] [Green Version]
- Pan, B.T.; Teng, K.; Wu, C.; Adam, M.; Johnstone, R.M. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J. Cell Boil. 1985, 101, 942–948. [Google Scholar] [CrossRef] [Green Version]
- Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nature Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef]
- Keerthikumar, S.; Gangoda, L.; Liem, M.; Fonseka, P.; Atukorala, I.; Ozcitti, C.; Mechler, A.I.; Adda, C.; Ang, C.-S.; Mathivanan, S. Proteogenomic analysis reveals exosomes are more oncogenic than ectosomes. Oncotarget 2015, 6, 15375–15396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- György, B.; Szabó, T.G.; Pásztói, M.; Pál, Z.; Misják, P.; Aradi, B.; László, V.; Pállinger, É.; Pap, E.; Kittel, Á.; et al. Membrane vesicles, current state-of-the-art: Emerging role of extracellular vesicles. Cell. Mol. Life Sci. 2011, 68, 2667–2688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Atkin-Smith, G.K.; Tixeira, R.; Paone, S.; Mathivanan, S.; Collins, C.; Liem, M.; Goodall, K.J.; Ravichandran, K.S.; Hulett, M.D.; Poon, I.K.H. A novel mechanism of generating extracellular vesicles during apoptosis via a beads-on-a-string membrane structure. Nat. Commun. 2015, 6, 7439. [Google Scholar] [CrossRef] [PubMed]
- Yuana, Y.; Sturk, A.; Nieuwland, R. Extracellular vesicles in physiological and pathological conditions. Blood Rev. 2013, 27, 31–39. [Google Scholar] [CrossRef] [Green Version]
- Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef]
- Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
- Margolis, L.; Sadovsky, Y. The biology of extracellular vesicles: The known unknowns. PLoS Boil. 2019, 17, e3000363. [Google Scholar] [CrossRef]
- Maacha, S.; Bhat, A.A.; Jimenez, L.; Raza, A.; Haris, M.; Uddin, S.; Grivel, J.-C. Extracellular vesicles-mediated intercellular communication: Roles in the tumor microenvironment and anti-cancer drug resistance. Mol. Cancer 2019, 18, 55. [Google Scholar] [CrossRef] [Green Version]
- Pathan, M.; Fonseka, P.; Chitti, S.V.; Kang, T.; Sanwlani, R.; Van Deun, J.; Hendrix, A.; Mathivanan, S. Vesiclepedia 2019: A compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 2018, 47, D516–D519. [Google Scholar] [CrossRef] [Green Version]
- Jin, Y.; Chen, K.; Wang, Z.; Wang, Y.; Liu, J.; Lin, L.; Shao, Y.; Gao, L.; Yin, H.; Cui, C.; et al. DNA in serum extracellular vesicles is stable under different storage conditions. BMC Cancer 2016, 16, 753. [Google Scholar] [CrossRef] [Green Version]
- Kahlert, C.; Melo, S.A.; Protopopov, A.; Tang, J.; Seth, S.; Koch, M.; Zhang, J.; Weitz, J.; Chin, L.; Futreal, A.; et al. Identification of Double-stranded Genomic DNA Spanning All Chromosomes with Mutated KRAS and p53 DNA in the Serum Exosomes of Patients with Pancreatic Cancer. J. Boil. Chem. 2014, 289, 3869–3875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, T.H.; Chennakrishnaiah, S.; Audemard, E.O.; Montermini, L.; Meehan, B.; Rak, J. Oncogenic ras-driven cancer cell vesiculation leads to emission of double-stranded DNA capable of interacting with target cells. Biochem. Biophys. Res. Commun. 2014, 451, 295–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thakur, B.K.; Zhang, H.; Becker, A.; Matei, I.; Huang, Y.; Costa-Silva, B.; Zheng, Y.; Hoshino, A.; Brazier, H.; Xiang, J.; et al. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 2014, 24, 766–769. [Google Scholar] [CrossRef] [Green Version]
- Boukouris, S.; Mathivanan, S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteom. Clin. Appl. 2015, 9, 358–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vagner, T.; Spinelli, C.; Minciacchi, V.R.; Balaj, L.; Zandian, M.; Conley, A.; Zijlstra, A.; Freeman, M.R.; Demichelis, F.; De, S.; et al. Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J. Extracell. Vesicles 2018, 7, 1505403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mathivanan, S.; Lim, J.W.E.; Tauro, B.J.; Ji, H.; Moritz, R.L.; Simpson, R. Proteomics analysis of A33 immunoaffinity-purified exosomes released from the human colon tumor cell line LIM1215 reveals a tissue-specific protein signature. Mol. Cell. Proteom. 2009, 9, 197–208. [Google Scholar] [CrossRef] [Green Version]
- Sun, J.-J.; Aswath, K.; Schroeder, S.G.; Lippolis, J.; Reinhardt, T.; Sonstegard, T.S. MicroRNA expression profiles of bovine milk exosomes in response to Staphylococcus aureus infection. BMC Genom. 2015, 16, 806. [Google Scholar] [CrossRef] [Green Version]
- Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti, N.; et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Boil. 2015, 428, 688–692. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, P.K. Interaction ofheatshockproteins withpeptides andantigenpresentingcells: Chaperoning of the Innate and Adaptive Immune Responses. Annu. Rev. Immunol. 2002, 20, 395–425. [Google Scholar] [CrossRef]
- Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef]
- Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef] [PubMed]
- Gangoda, L.; Boukouris, S.; Liem, M.; Kalra, H.; Mathivanan, S. Extracellular vesicles including exosomes are mediators of signal transduction: Are they protective or pathogenic? Proteomics 2014, 15, 260–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, D.; Kim, D.-K.; Kim, Y.-K.; Gho, Y.S. Proteomics of extracellular vesicles: Exosomes and ectosomes. Mass Spectrom. Rev. 2014, 34, 474–490. [Google Scholar] [CrossRef] [PubMed]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
- Turchinovich, A.; Drapkina, O.; Tonevitsky, A. Transcriptome of Extracellular Vesicles: State-of-the-Art. Front. Immunol. 2019, 10, 202. [Google Scholar] [CrossRef] [Green Version]
- Mulcahy, L.; Pink, R.C.; Carter, D.R.F. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 1093. [Google Scholar] [CrossRef] [Green Version]
- Llorente, A.; Skotland, T.; Sylvänne, T.; Kauhanen, D.; Rog, T.; Orłowski, A.; Vattulainen, I.; Ekroos, K.; Sandvig, K. Molecular lipidomics of exosomes released by PC-3 prostate cancer cells. Biochim. Biophys. Acta (BBA) - Bioenerg. 2013, 1831, 1302–1309. [Google Scholar] [CrossRef]
- Skotland, T.; Sandvig, K.; Llorente, A. Lipids in exosomes: Current knowledge and the way forward. Prog. Lipid Res. 2017, 66, 30–41. [Google Scholar] [CrossRef]
- Lydic, T.A.; Townsend, S.; Adda, C.; Collins, C.; Mathivanan, S.; Reid, G. Rapid and comprehensive ’shotgun’ lipidome profiling of colorectal cancer cell derived exosomes. Methods 2015, 87, 83–95. [Google Scholar] [CrossRef] [Green Version]
- Kim, C.W.; Lee, H.M.; Lee, T.H.; Kang, C.; Kleinman, H.K.; Gho, Y.S. Extracellular membrane vesicles from tumor cells promote angiogenesis via sphingomyelin. Cancer Res. 2002, 62, 6312–6317. [Google Scholar]
- Svenningsen, P.; Sabaratnam, R.; Jensen, B.L. Urinary extracellular vesicles: Origin, role as intercellular messengers and biomarkers; efficient sorting and potential treatment options. Acta Physiol. 2019, 228, e13346. [Google Scholar] [CrossRef] [PubMed]
- Admyre, C.; Johansson, S.M.; Qazi, K.R.; Filén, J.-J.; Lahesmaa, R.; Norman, M.; Neve, E.P.A.; Scheynius, A.; Gabrielsson, S. Exosomes with immune modulatory features are present in human breast milk. J. Immunol. 2007, 179, 1969–1978. [Google Scholar] [CrossRef] [PubMed]
- Antounians, L.; Tzanetakis, A.; Pellerito, O.; Catania, V.D.; Sulistyo, A.; Montalva, L.; McVey, M.J.; Zani, A. The Regenerative Potential of Amniotic Fluid Stem Cell Extracellular Vesicles: Lessons Learned by Comparing Different Isolation Techniques. Sci. Rep. 2019, 9, 1837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Welton, J.L.; Loveless, S.; Stone, T.; Von Ruhland, C.; Robertson, N.P.; Clayton, A. Cerebrospinal fluid extracellular vesicle enrichment for protein biomarker discovery in neurological disease; multiple sclerosis. J. Extracell. Vesicles 2017, 6, 1369805. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Jia, L.; Zheng, Y.; Li, W. Salivary Exosomes: Emerging Roles in Systemic Disease. Int. J. Boil. Sci. 2018, 14, 633–643. [Google Scholar] [CrossRef]
- Fernando, M.R.; Jiang, C.; Krzyzanowski, G.D.; Ryan, W.L. New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS ONE 2017, 12, e0183915. [Google Scholar] [CrossRef] [Green Version]
- Kalra, H.; Gangoda, L.; Fonseka, P.; Chitti, S.V.; Liem, M.; Keerthikumar, S.; Samuel, M.; Boukouris, S.; Al Saffar, H.; Collins, C.; et al. Extracellular vesicles containing oncogenic mutant β-catenin activate Wnt signalling pathway in the recipient cells. J. Extracell. Vesicles 2019, 8, 1690217. [Google Scholar] [CrossRef] [Green Version]
- Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Mark, M.T.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
- Yuyama, K.; Sun, H.; Sakai, S.; Mitsutake, S.; Okada, M.; Tahara, H.; Furukawa, J.-I.; Fujitani, N.; Shinohara, Y.; Igarashi, Y. Decreased Amyloid-β Pathologies by Intracerebral Loading of Glycosphingolipid-enriched Exosomes in Alzheimer Model Mice. J. Boil. Chem. 2014, 289, 24488–24498. [Google Scholar] [CrossRef] [Green Version]
- Yuyama, K.; Sun, H.; Usuki, S.; Sakai, S.; Hanamatsu, H.; Mioka, T.; Kimura, N.; Okada, M.; Tahara, H.; Furukawa, J.-I.; et al. A potential function for neuronal exosomes: Sequestering intracerebral amyloid-β peptide. FEBS Lett. 2014, 589, 84–88. [Google Scholar] [CrossRef]
- Barclay, R.A.; Schwab, A.; DeMarino, C.; Akpamagbo, Y.; Lepene, B.; Kassaye, S.; Iordanskiy, S.; Kashanchi, F. Exosomes from uninfected cells activate transcription of latent HIV-1. J. Boil. Chem. 2017, 292, 11682–11701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mori, Y.; Koike, M.; Moriishi, E.; Kawabata, A.; Tang, H.; Oyaizu, H.; Uchiyama, Y.; Yamanishi, K. Human Herpesvirus-6 Induces MVB Formation, and Virus Egress Occurs by an Exosomal Release Pathway. Traffic 2008, 9, 1728–1742. [Google Scholar] [CrossRef] [Green Version]
- Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef] [PubMed]
- Janowska-Wieczorek, A.; Wysoczynski, M.; Kijowski, J.; Marquez-Curtis, L.; Machalinski, B.; Ratajczak, J.; Ratajczak, M.Z. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Int. J. Cancer 2004, 113, 752–760. [Google Scholar] [CrossRef]
- Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [Green Version]
- Tominaga, N.; Kosaka, N.; Ono, M.; Katsuda, T.; Yoshioka, Y.; Tamura, K.; Lötvall, J.; Nakagama, H.; Ochiya, T. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood–brain barrier. Nat. Commun. 2015, 6, 6716. [Google Scholar] [CrossRef] [Green Version]
- Hong, Y.; Zhao, T.; Li, X.-J.; Li, S. Mutant Huntingtin Inhibits αB-Crystallin Expression and Impairs Exosome Secretion from Astrocytes. J. Neurosci. 2017, 37, 9550–9563. [Google Scholar] [CrossRef] [Green Version]
- Peng, K.Y.; Perez-Gonzalez, R.; Alldred, M.J.; Goulbourne, C.N.; Morales-Corraliza, J.; Saito, M.; Saito, M.; Ginsberg, S.D.; Mathews, P.; Levy, E. Apolipoprotein E4 genotype compromises brain exosome production. Brain 2018, 142, 163–175. [Google Scholar] [CrossRef] [Green Version]
- Panigrahi, G.; Praharaj, P.P.; Peak, T.C.; Long, J.; Singh, R.; Rhim, J.S.; Elmageed, Z.Y.A.; Deep, G. Hypoxia-induced exosome secretion promotes survival of African-American and Caucasian prostate cancer cells. Sci. Rep. 2018, 8, 3853. [Google Scholar] [CrossRef]
- Keklikoglou, I.; Cianciaruso, C.; Güç, E.; Squadrito, M.L.; Spring, L.; Tazzyman, S.; Lambein, L.; Poissonnier, A.; Ferraro, G.B.; Baer, C.; et al. Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models. Nature 2018, 21, 190–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, G.; Liu, Z.; Ding, H.; Zhou, Y.; Doan, H.A.; Sin, K.W.T.; Zhu, Z.J.; Flores, R.; Wen, Y.; Gong, X.; et al. Tumor induces muscle wasting in mice through releasing extracellular Hsp70 and Hsp. Nat. Commun. 2017, 8, 589. [Google Scholar] [CrossRef] [Green Version]
- Chitti, S.V.; Fonseka, P.; Mathivanan, S. Emerging role of extracellular vesicles in mediating cancer cachexia. Biochem. Soc. Trans. 2018, 46, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
- Fonseka, P.; Liem, M.; Ozcitti, C.; Adda, C.G.; Ang, C.-S.; Mathivanan, S. Exosomes from N-Myc amplified neuroblastoma cells induce migration and confer chemoresistance to non-N-Myc amplified cells: Implications of intra-tumour heterogeneity. J. Extracell. Vesicles 2019, 8, 1597614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bandari, S.K.; Purushothaman, A.; Ramani, V.C.; Brinkley, G.J.; Chandrashekar, D.S.; Varambally, S.; Mobley, J.A.; Zhang, Y.; Brown, E.E.; Vlodavsky, I.; et al. Chemotherapy induces secretion of exosomes loaded with heparanase that degrades extracellular matrix and impacts tumor and host cell behavior. Matrix Boil. 2017, 65, 104–118. [Google Scholar] [CrossRef] [PubMed]
- Samuel, M.; Chisanga, D.; Liem, M.; Keerthikumar, S.; Anand, S.; Ang, C.-S.; Adda, C.G.; Versteegen, E.; Jois, M.; Mathivanan, S. Bovine milk-derived exosomes from colostrum are enriched with proteins implicated in immune response and growth. Sci. Rep. 2017, 7, 5933. [Google Scholar] [CrossRef]
- Hosseini, S.M.; Talaei-Khozani, T.; Sani, M.; Owrangi, B. Differentiation of Human Breast-Milk Stem Cells to Neural Stem Cells and Neurons. Neurol. Res. Int. 2014, 2014, 1–8. [Google Scholar] [CrossRef]
- Benmoussa, A.; Provost, P. Milk MicroRNAs in Health and Disease. Compr. Rev. Food Sci. Food Saf. 2019, 18, 703–722. [Google Scholar] [CrossRef] [Green Version]
- Xu, R.; Sangild, P.; Zhang, Y.; Zhang, S. Chapter 5 Bioactive compounds in porcine colostrum and milk and their effects on intestinal development in neonatal pigs. In Biology of the Intestine in Growing Animals; Zabielski, R., Gregory, P.C., Weström, B., Salek, E., Eds.; Elsevier BV: Amsterdam, The Netherlands, 2002; Volume 1, pp. 169–192. [Google Scholar]
- Tripathi, V.; Vashishtha, B. Bioactive Compounds of Colostrum and Its Application. Food Rev. Int. 2006, 22, 225–244. [Google Scholar] [CrossRef]
- Burns, T. Colostrum. In Current Therapy in Equine Reproduction; Samper, J.C., Pycock, J.F., McKinnon, A.O., Eds.; W.B. Saunders: Saint Louis, MO, USA, 2007; pp. 452–453. [Google Scholar] [CrossRef]
- Ballard, O.; Morrow, A.L. Human milk composition: Nutrients and bioactive factors. Pediatr. Clin. North. Am. 2013, 60, 49–74. [Google Scholar] [CrossRef] [Green Version]
- Le Doare, K.; Holder, B.; Bassett, A.; Pannaraj, P.S. Mother’s Milk: A Purposeful Contribution to the Development of the Infant Microbiota and Immunity. Front. Immunol. 2018, 9, 361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- German, J.B.; Dillard, C.J.; Ward, R.E. Bioactive components in milk. Curr. Opin. Clin. Nutr. Metab. Care 2002, 5, 653–658. [Google Scholar] [CrossRef] [PubMed]
- Chen, P.-W.; Lin, Y.-L.; Huang, M.-S. Profiles of commensal and opportunistic bacteria in human milk from healthy donors in Taiwan. J. Food Drug Anal. 2018, 26, 1235–1244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martín, R.; Langa, S.; Reviriego, C.; Jiménez, E.; Marin, M.L.; Xaus, J.; Fernández, L.; Rodríguez, J.M. Human milk is a source of lactic acid bacteria for the infant gut. J. Pediatr. 2003, 143, 754–758. [Google Scholar] [CrossRef]
- Witkowska-Zimny, M.; Kaminska-El-Hassan, E. Cells of human breast milk. Cell. Mol. Boil. Lett. 2017, 22, 11. [Google Scholar] [CrossRef]
- Ninkina, N.; Kukharsky, M.S.; Hewitt, M.V.; Lysikova, E.A.; Skuratovska, L.N.; Deykin, A.; Buchman, V.L. Stem cells in human breast milk. Hum. Cell 2019, 32, 223–230. [Google Scholar] [CrossRef] [Green Version]
- Melnik, B.C.; John, S.; Schmitz, G. Milk is not just food but most likely a genetic transfection system activating mTORC1 signaling for postnatal growth. Nutr. J. 2013, 12, 103. [Google Scholar] [CrossRef] [Green Version]
- Andreas, N.J.; Kampmann, B.; Le Doare, K. Human breast milk: A review on its composition and bioactivity. Early Hum. Dev. 2015, 91, 629–635. [Google Scholar] [CrossRef]
- Pieters, B.C.H.; Arntz, O.J.; Bennink, M.B.; Broeren, M.; Van Caam, A.; Koenders, M.I.; Van Lent, P.L.E.M.; Berg, W.B.V.D.; De Vries, M.; Van Der Kraan, P.M.; et al. Commercial Cow Milk Contains Physically Stable Extracellular Vesicles Expressing Immunoregulatory TGF-β. PLoS ONE 2015, 10, e0121123. [Google Scholar] [CrossRef]
- Baier, S.R.; Nguyen, C.; Xie, F.; Wood, J.R.; Zempleni, J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J. Nutr. 2014, 144, 1495–1500. [Google Scholar] [CrossRef]
- Melnik, B.C.; Schmitz, G. Milk’s Role as an Epigenetic Regulator in Health and Disease. Diseases 2017, 5, 12. [Google Scholar] [CrossRef] [PubMed]
- Kalliomäki, M.; Ouwehand, A.C.; Arvilommi, H.; Kero, P.; Isolauri, E. Transforming growth factor-β in breast milk: A potential regulator of atopic disease at an early age. J. Allergy Clin. Immunol. 1999, 104, 1251–1257. [Google Scholar] [CrossRef]
- Okuda, M.; Miyashiro, E.; Nakazawa, T.; Yamauchi, K.; Koizumi, R.; Teraguchi, S.; Tamura, Y.; Booka, M.; Yoshikawa, N.; Adachi, Y.; et al. Bovine lactoferrin is effective to suppress Helicobacter pylori colonization in the human stomach: A randomized, double-blind, placebo-controlled study. J. Infect. Chemother. 2005, 11, 265–269. [Google Scholar] [CrossRef] [PubMed]
- McIntosh, G.H.; Wang, Y.H.A.; Royle, P.J. A diet containing chickpeas and wheat offers less protection against colon tumors than a casein and wheat diet in dimethylhydrazine-treated rats. J. Nutr. 1998, 128, 804–809. [Google Scholar] [CrossRef] [Green Version]
- Ford, J.T.; Wong, C.W.; Colditz, I.G. Effects of dietary protein types on immune responses and levels of infection withEimeria vermiformisin mice. Immunol. Cell Boil. 2001, 79, 23–28. [Google Scholar] [CrossRef]
- Davoodi, S.H.; Shahbazi, R.; Esmaeili, S.; Sohrabvandi, S.; Mortazavian, A.; Jazayeri, S.; Taslimi, A. Health-Related Aspects of Milk Proteins. Iran. J. Pharm. Res. IJPR 2016, 15, 573–591. [Google Scholar]
- Wiley, A.S. Cow milk consumption, insulin-like growth factor-I, and human biology: A life history approach. Am. J. Hum. Boil. 2011, 24, 130–138. [Google Scholar] [CrossRef]
- Melnik, B.C.; Schmitz, G. Exosomes of pasteurized milk: Potential pathogens of Western diseases. J. Transl. Med. 2019, 17, 3. [Google Scholar] [CrossRef] [Green Version]
- Melnik, B.C.; Schmitz, G. DNA methyltransferase 1-targeting miRNA-148a of dairy milk: A potential bioactive modifier of the human epigenome. Funct. Foods Heal. Dis. 2017, 7, 671. [Google Scholar] [CrossRef] [Green Version]
- Samuel, M.; Bleackley, M.R.; Anderson, M.A.; Mathivanan, S. Extracellular vesicles including exosomes in cross kingdom regulation: A viewpoint from plant-fungal interactions. Front. Plant. Sci. 2015, 6, 1695. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Hou, D.; Chen, X.; Li, D.; Zhu, L.; Zhang, Y.; Li, J.; Bian, Z.; Liang, X.; Cai, X.; et al. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: Evidence of cross-kingdom regulation by microRNA. Cell Res. 2012, 22, 107–126. [Google Scholar] [CrossRef] [PubMed]
- Benmoussa, A.; Lee, C.H.C.; Laffont, B.; Savard, P.; Laugier, J.; Boilard, E.; Gilbert, C.; Fliss, I.; Provost, P. Commercial Dairy Cow Milk microRNAs Resist Digestion under Simulated Gastrointestinal Tract Conditions. J. Nutr. 2016, 146, 2206–2215. [Google Scholar] [CrossRef]
- Link, J.; Thon, C.; Schanze, D.; Steponaitiene, R.; Kupcinskas, J.; Zenker, M.; Canbay, A.; Malfertheiner, P.; Link, A. Food-Derived Xeno-microRNAs: Influence of Diet and Detectability in Gastrointestinal Tract—Proof-of-Principle Study. Mol. Nutr. Food Res. 2018, 63, 1800076. [Google Scholar] [CrossRef] [PubMed]
- 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, 4831–4841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gupta, S.; Haigh, B.J.; Seyfert, H.M.; Griffin, F.; Wheeler, T.T. Bovine milk RNases modulate pro-inflammatory responses induced by nucleic acids in cultured immune and epithelial cells. Dev. Comp. Immunol. 2017, 68, 87–97. [Google Scholar] [CrossRef] [PubMed]
- Witwer, K.W.; McAlexander, M.A.; Queen, S.E.; Adams, R.J. Real-time quantitative PCR and droplet digital PCR for plant miRNAs in mammalian blood provide little evidence for general uptake of dietary miRNAs: Limited evidence for general uptake of dietary plant xenomiRs. RNA Boil. 2013, 10, 1080–1086. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Zhang, Y.; Dong, P.; An, R.; Xue, C.; Ge, Y.; Wei, L.; Liang, X. Digestion of Nucleic Acids Starts in the Stomach. Sci. Rep. 2015, 5, 11936. [Google Scholar] [CrossRef] [Green Version]
- Tomé-Carneiro, J.; Fernández-Alonso, N.; Tomás-Zapico, C.; Visioli, F.; Iglesias-Gutierrez, E.; Davalos, A. Breast milk microRNAs harsh journey towards potential effects in infant development and maturation. Lipid encapsulation can help. Pharmacol. Res. 2018, 132, 21–32. [Google Scholar] [CrossRef]
- Benmoussa, A.; Ly, S.; Shan, S.T.; Laugier, J.; Boilard, E.; Gilbert, C.; Provost, P. A subset of extracellular vesicles carries the bulk of microRNAs in commercial dairy cow’s milk. J. Extracell. Vesicles 2017, 6, 1401897. [Google Scholar] [CrossRef] [Green Version]
- Izumi, H.; Tsuda, M.; Sato, Y.; Kosaka, N.; Ochiya, T.; Iwamoto, H.; Namba, K.; Takeda, Y. Bovine milk exosomes contain microRNA and mRNA and are taken up by human macrophages. J. Dairy Sci. 2015, 98, 2920–2933. [Google Scholar] [CrossRef] [Green Version]
- Manca, S.; Upadhyaya, B.; Mutai, E.; Desaulniers, A.T.; Cederberg, R.A.; White, B.; Zempleni, J. Milk exosomes are bioavailable and distinct microRNA cargos have unique tissue distribution patterns. Sci. Rep. 2018, 8, 11321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baddela, V.S.; Nayan, V.; Rani, P.; Onteru, S.K.; Singh, D. Physicochemical Biomolecular Insights into Buffalo Milk-Derived Nanovesicles. Appl. Biochem. Biotechnol. 2015, 178, 544–557. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.; Xie, M.-Y.; Sun, J.-J.; Ye, R.-S.; Cheng, X.; Sun, R.-P.; Wei, L.-M.; Li, M.; Lin, D.-L.; Jiang, Q.-Y.; et al. Porcine milk-derived exosomes promote proliferation of intestinal epithelial cells. Sci. Rep. 2016, 6, 33862. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.; Xi, Q.-Y.; Sun, J.-J.; Ye, R.-S.; Cheng, X.; Sun, R.-P.; Wang, S.; Shu, G.; Wang, L.-N.; Zhu, X.; et al. Revelation of mRNAs and proteins in porcine milk exosomes by transcriptomic and proteomic analysis. BMC Veter- Res. 2017, 13, 101. [Google Scholar] [CrossRef] [PubMed]
- Sedykh, S.E.; Purvinish, L.V.; Monogarov, A.S.; Burkova, E.E.; Grigor’Eva, A.E.; Bulgakov, D.V.; Dmitrenok, P.S.; Vlassov, V.V.; Ryabchikova, E.I.; Nevinsky, G.A. Purified horse milk exosomes contain an unpredictable small number of major proteins. Biochim. Open 2017, 4, 61–72. [Google Scholar] [CrossRef] [PubMed]
- Reinhardt, T.; Lippolis, J.D.; Nonnecke, B.J.; Sacco, R.E. Bovine milk exosome proteome. J. Proteom. 2012, 75, 1486–1492. [Google Scholar] [CrossRef]
- Benmoussa, A.; Gotti, C.; Bourassa, S.; Gilbert, C.; Provost, P. Identification of protein markers for extracellular vesicle (EV) subsets in cow’s milk. J. Proteom. 2019, 192, 78–88. [Google Scholar] [CrossRef]
- Zhou, Q.; Li, M.; Wang, X.; Li, Q.; Wang, T.; Zhu, Q.; Zhou, X.; Wang, X.; Gao, X.; Li, X. Immune-related MicroRNAs are Abundant in Breast Milk Exosomes. Int. J. Boil. Sci. 2011, 8, 118–123. [Google Scholar] [CrossRef]
- Zempleni, J.; Aguilar-Lozano, A.; Sadri, M.; Sukreet, S.; Manca, S.; Wu, D.; Zhou, F.; Mutai, E. Biological Activities of Extracellular Vesicles and Their Cargos from Bovine and Human Milk in Humans and Implications for Infants. J. Nutr. 2016, 147, 3–10. [Google Scholar] [CrossRef] [Green Version]
- Hassiotou, F.; Beltran, A.; Chetwynd, E.; Stuebe, A.M.; Twigger, A.-J.; Metzger, P.; Trengove, N.; Lai, C.T.; Filgueira, L.; Blancafort, P.; et al. Breastmilk Is a Novel Source of Stem Cells with Multilineage Differentiation Potential. STEM CELLS 2012, 30, 2164–2174. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.; Shimizu, K.; Yamauchi, M.; Takase, H.; Ugawa, S.; Okada, A.; Inoshima, Y. Acidification effects on isolation of extracellular vesicles from bovine milk. PLoS ONE 2019, 14, e0222613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wolf, T.; Baier, S.R.; Zempleni, J. The Intestinal Transport of Bovine Milk Exosomes Is Mediated by Endocytosis in Human Colon Carcinoma Caco-2 Cells and Rat Small Intestinal IEC-6 Cells. J. Nutr. 2015, 145, 2201–2206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berk, M.; Williams, L.J.; Jacka, F.; O’Neil, A.; Pasco, J.; Moylan, S.; Allen, N.B.; Stuart, A.L.; Hayley, A.C.; Byrne, M.L.; et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med. 2013, 11, 200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tulkens, J.; Vergauwen, G.; Van Deun, J.; Geeurickx, E.; Dhondt, B.; Lippens, L.; De Scheerder, M.-A.; Miinalainen, I.; Rappu, P.; De Geest, B.G.; et al. Increased levels of systemic LPS-positive bacterial extracellular vesicles in patients with intestinal barrier dysfunction. Gut 2018, 69, 191–193. [Google Scholar] [CrossRef] [Green Version]
- Reif, S.; Shiff, Y.E.; Golan-Gerstl, R. Milk-derived exosomes (MDEs) have a different biological effect on normal fetal colon epithelial cells compared to colon tumor cells in a miRNA-dependent manner. J. Transl. Med. 2019, 17, 325. [Google Scholar] [CrossRef]
- Messica, A.; Mehrez, A. Time-to-market, window of opportunity, and salvageability of a new product development. Manag. Decis. Econ. 2002, 23, 371–378. [Google Scholar] [CrossRef]
- Kusuma, R.J.; Manca, S.; Friemel, T.; Sukreet, S.; Nguyen, C.; Zempleni, J. Human vascular endothelial cells transport foreign exosomes from cow’s milk by endocytosis. Am. J. Physiol. Physiol. 2016, 310, C800–C807. [Google Scholar] [CrossRef] [Green Version]
- Howard, K.M.; Kusuma, R.J.; Baier, S.R.; Friemel, T.; Markham, L.; Vanamala, J.; Zempleni, J.; Zempleni, J. Loss of miRNAs during Processing and Storage of Cow’s (Bos taurus) Milk. J. Agric. Food Chem. 2015, 63, 588–592. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Z.; Yu, S.; Xu, M.; Li, P. Effects of microwave on extracellular vesicles and microRNA in milk. J. Dairy Sci. 2018, 101, 2932–2940. [Google Scholar] [CrossRef]
- Kirchner, B.; Pfaffl, M.W.; Dumpler, J.; Von Mutius, E.; Ege, M.J. microRNA in native and processed cow’s milk and its implication for the farm milk effect on asthma. J. Allergy Clin. Immunol. 2016, 137, 1893–1895.e13. [Google Scholar] [CrossRef] [Green Version]
- van Herwijnen, M.J.; Zonneveld, M.I.; Goerdayal, S.; Nolte, E.N.; Garssen, J.; Stahl, B.; Altelaar, A.M.; Redegeld, F.A.; Wauben, M.H. Comprehensive Proteomic Analysis of Human Milk-derived Extracellular Vesicles Unveils a Novel Functional Proteome Distinct from Other Milk Components. Mol. Cell. Proteom. 2016, 15, 3412–3423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vaswani, K.; Koh, Y.Q.; Almughlliq, F.; Peiris, H.; Mitchell, M. A method for the isolation and enrichment of purified bovine milk exosomes. Reprod. Boil. 2017, 17, 341–348. [Google Scholar] [CrossRef] [PubMed]
- Benmoussa, A.; Laugier, J.; Beauparlant, C.J.; Lambert, M.; Droit, A.; Provost, P. Complexity of the microRNA transcriptome of cow milk and milk-derived extracellular vesicles isolated via differential ultracentrifugation. J. Dairy Sci. 2019, 103, 16–29. [Google Scholar] [CrossRef]
- Qin, W.; Tsukasaki, Y.; Dasgupta, S.; Mukhopadhyay, N.; Ikebe, M.; Sauter, E.R. Exosomes in Human Breast Milk Promote EMT. Clin. Cancer Res. 2016, 22, 4517–4524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Melnik, B.C.; John, S.; Carrera-Bastos, P.; Schmitz, G. Milk: A postnatal imprinting system stabilizing FoxP3 expression and regulatory T cell differentiation. Clin. Transl. Allergy 2016, 6, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oliveira, M.C.; Arntz, O.J.; Davidson, E.N.B.; Van Lent, P.L.; Koenders, M.; Van Der Kraan, P.; Berg, W.B.V.D.; Ferreira, A.V.M.; Van De Loo, F.A.; Information, P.E.K.F.C. Milk extracellular vesicles accelerate osteoblastogenesis but impair bone matrix formation. J. Nutr. Biochem. 2016, 30, 74–84. [Google Scholar] [CrossRef] [PubMed]
- Liao, Y.; Alvarado, R.; Phinney, B.; Lönnerdal, B. Proteomic Characterization of Human Milk Whey Proteins during a Twelve-Month Lactation Period. J. Proteome Res. 2011, 10, 1746–1754. [Google Scholar] [CrossRef]
- Reinhardt, T.; Sacco, R.E.; Nonnecke, B.; Lippolis, J. Bovine milk proteome: Quantitative changes in normal milk exosomes, milk fat globule membranes and whey proteomes resulting from Staphylococcus aureus mastitis. J. Proteom. 2013, 82, 141–154. [Google Scholar] [CrossRef]
- Melnik, B.C.; Kakulas, F.; Patel, V.; Preedy, V. Milk Exosomes and microRNAs: Potential Epigenetic Regulators. In Handbook of Nutrition, Diet, and Epigenetics; Patel, V., Preedy, V., Eds.; Springer Nature Switzerland AG: Cham, Switzerland, 2017; pp. 1–28. [Google Scholar]
- Sun, Q.; Chen, X.; Yu, J.; Zen, K.; Zhang, C.; Li, L. Immune modulatory function of abundant immune-related microRNAs in microvesicles from bovine colostrum. Protein Cell 2013, 4, 197–210. [Google Scholar] [CrossRef] [Green Version]
- Colitti, M.; Sgorlon, S.; Licastro, D.; Stefanon, B. Differential expression of miRNAs in milk exosomes of cows subjected to group relocation. Res. Veter- Sci. 2019, 122, 148–155. [Google Scholar] [CrossRef]
- Mobley, C.B.; Mumford, P.W.; McCarthy, J.J.; Miller, M.E.; Young, K.C.; Martin, J.S.; Beck, D.; Lockwood, C.; Roberts, M.D. Whey protein-derived exosomes increase protein synthesis and hypertrophy in C2C12 myotubes. J. Dairy Sci. 2017, 100, 48–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leiferman, A.; Shu, J.; Grove, R.; Cui, J.; Adamec, J.; Zempleni, J. A diet defined by its content of bovine milk exosomes and their RNA cargos has moderate effects on gene expression, amino acid profiles and grip strength in skeletal muscle in C57BL/6 mice. J. Nutr. Biochem. 2018, 59, 123–128. [Google Scholar] [CrossRef] [PubMed]
- Martin, C.; Patel, M.; Williams, S.; Arora, H.; Sims, B.; Brawner, K. Human breast milk-derived exosomes attenuate cell death in intestinal epithelial cells. Innate Immun. 2018, 24, 278–284. [Google Scholar] [CrossRef] [PubMed]
- Melnik, B.C.; John, S.; Schmitz, G. Milk: An exosomal microRNA transmitter promoting thymic regulatory T cell maturation preventing the development of atopy? J. Transl. Med. 2014, 12, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, X.; Gao, C.; Li, H.; Huang, L.; Sun, Q.; Dong, Y.; Tian, C.; Gao, S.; Dong, H.; Guan, D.; 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, 1128–1137. [Google Scholar] [CrossRef]
- Melnik, B.C. Milk: An epigenetic amplifier of FTO-mediated transcription? Implications for Western diseases. J. Transl. Med. 2015, 13, 385. [Google Scholar] [CrossRef] [Green Version]
- Melnik, B.C. Milk—A Nutrient System of Mammalian Evolution Promoting mTORC1-Dependent Translation. Int. J. Mol. Sci. 2015, 16, 17048–17087. [Google Scholar] [CrossRef]
- Boissel, S.; Reish, O.; Proulx, K.; Kawagoe-Takaki, H.; Sedgwick, B.; Yeo, G.S.H.; Meyre, D.; Golzio, C.; Molinari, F.; Kadhom, N.; et al. Loss-of-Function Mutation in the Dioxygenase-Encoding FTO Gene Causes Severe Growth Retardation and Multiple Malformations. Am. J. Hum. Genet. 2009, 85, 106–111. [Google Scholar] [CrossRef] [Green Version]
- Fischer, J.; Koch, L.; Emmerling, C.; Vierkotten, J.; Peters, T.; Brüning, J.C.; Ruther, U. Inactivation of the Fto gene protects from obesity. Nature 2009, 458, 894–898. [Google Scholar] [CrossRef]
- Kuroda, A.; Rauch, T.A.; Todorov, I.; Ku, H.T.; Al-Abdullah, I.H.; Kandeel, F.; Mullen, Y.; Pfeifer, G.P.; Ferreri, K. Insulin gene expression is regulated by DNA methylation. PLoS ONE 2009, 4, e6953. [Google Scholar] [CrossRef] [Green Version]
- Dooley, J.; Garcia-Perez, J.E.; Sreenivasan, J.; Schlenner, S.; Vangoitsenhoven, R.; Papadopoulou, A.S.; Tian, L.; Schonefeldt, S.; Serneels, L.; Deroose, C.; et al. The microRNA-29 Family Dictates the Balance Between Homeostatic and Pathological Glucose Handling in Diabetes and Obesity. Diabetes 2015, 65, 53–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoppe, C.; Mølgaard, C.; Michaelsen, K.F. Cow’s Milk and Linear Growth in Industrialized and Developing Countries. Annu. Rev. Nutr. 2006, 26, 131–173. [Google Scholar] [CrossRef] [PubMed]
- Qin, L.; He, K.; Xu, J.-Y. Milk consumption and circulating insulin-like growth factor-I level: A systematic literature review. Int. J. Food Sci. Nutr. 2009, 60, 330–340. [Google Scholar] [CrossRef] [PubMed]
- Benmoussa, A.; Diallo, I.; Salem, M.; Michel, S.; Gilbert, C.; Sévigny, J.; Provost, P. Concentrates of two subsets of extracellular vesicles from cow’s milk modulate symptoms and inflammation in experimental colitis. Sci. Rep. 2019, 9, 14661–14716. [Google Scholar] [CrossRef] [Green Version]
- Palomares, O.; Yaman, G.; Azkur, A.K.; Akkoc, T.; Akdis, M.; Akdis, C.A. Role of Treg in immune regulation of allergic diseases. Eur. J. Immunol. 2010, 40, 1232–1240. [Google Scholar] [CrossRef]
- Neudecker, V.; Haneklaus, M.; Jensen, O.; Khailova, L.; Masterson, J.C.; Tye, H.; Biette, K.; Jedlicka, P.; Brodsky, K.S.; Gerich, M.E.; et al. Myeloid-derived miR-223 regulates intestinal inflammation via repression of the NLRP3 inflammasome. J. Exp. Med. 2017, 214, 1737–1752. [Google Scholar] [CrossRef]
- Liao, Y.; Du, X.; Li, J.; Lönnerdal, B. Human milk exosomes and their microRNAs survive digestion in vitro and are taken up by human intestinal cells. Mol. Nutr. Food Res. 2017, 61, 1700082. [Google Scholar] [CrossRef]
- Karlsson, O.; Rodosthenous, R.; Jara, C.; Brennan, K.J.; Wright, R.O.; Baccarelli, A.A.; Wright, R.J. Detection of long non-coding RNAs in human breastmilk extracellular vesicles: Implications for early child development. Epigenetics 2016, 11, 721–729. [Google Scholar] [CrossRef] [Green Version]
- An, X.; Ma, K.; Zhang, Z.; Zhao, T.; Zhang, X.; Tang, B.; Li, Z. miR-17, miR-21, and miR-143 Enhance Adipogenic Differentiation from Porcine Bone Marrow-Derived Mesenchymal Stem Cells. DNA Cell Boil. 2016, 35, 410–416. [Google Scholar] [CrossRef]
- Zhang, J.; Ying, Z.-Z.; Tang, Z.; Long, L.-Q.; Li, K. MicroRNA-148a Promotes Myogenic Differentiation by Targeting the ROCK1 Gene*. J. Boil. Chem. 2012, 287, 21093–21101. [Google Scholar] [CrossRef] [Green Version]
- Noer, A.; Sørensen, A.L.; Boquest, A.C.; Collas, P. Stable CpG Hypomethylation of Adipogenic Promoters in Freshly Isolated, Cultured, and Differentiated Mesenchymal Stem Cells from Adipose Tissue. Mol. Boil. Cell 2006, 17, 3543–3556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parry, H.A.; Mobley, C.B.; Mumford, P.W.; Romero, M.A.; Haun, C.T.; Zhang, Y.; Roberson, P.A.; Zempleni, J.; Ferrando, A.A.; Vechetti-Junior, I.; et al. Bovine Milk Extracellular Vesicles (EVs) Modification Elicits Skeletal Muscle Growth in Rats. Front. Physiol. 2019, 10, 436. [Google Scholar] [CrossRef] [PubMed]
- Oliveira, M.C.; Di Ceglie, I.; Arntz, O.J.; Berg, W.B.V.D.; Hoogen, F.H.J.V.D.; Ferreira, A.V.M.; Van Lent, P.L.; Van De Loo, F.A. Milk-Derived Nanoparticle Fraction Promotes the Formation of Small Osteoclasts But Reduces Bone Resorption. J. Cell. Physiol. 2016, 232, 225–233. [Google Scholar] [CrossRef] [PubMed]
- Gao, H.; Guo, H.; Zhang, H.; Xie, X.; Wen, P.; Ren, F. Yak-milk-derived exosomes promote proliferation of intestinal epithelial cells in an hypoxic environment. J. Dairy Sci. 2019, 102, 985–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bian, Y.; Lei, Y.; Wang, C.; Wang, J.; Wang, L.; Liu, L.; Liu, L.; Gao, X.; Li, Q.-Z. Epigenetic Regulation of miR-29s Affects the Lactation Activity of Dairy Cow Mammary Epithelial Cells. J. Cell. Physiol. 2015, 230, 2152–2163. [Google Scholar] [CrossRef]
- Näslund, T.I.; Paquin-Proulx, D.; Paredes, P.T.; Vallhov, H.; Sandberg, J.K.; Gabrielsson, S. Exosomes from breast milk inhibit HIV-1 infection of dendritic cells and subsequent viral transfer to CD4+ T cells. AIDS 2014, 28, 171–180. [Google Scholar] [CrossRef] [Green Version]
- Saeland, E.; De Jong, M.A.; Nabatov, A.A.; Kalay, H.; Geijtenbeek, T.B.; Van Kooyk, Y. MUC1 in human milk blocks transmission of human immunodeficiency virus from dendritic cells to T cells. Mol. Immunol. 2009, 46, 2309–2316. [Google Scholar] [CrossRef]
- Naarding, M.A.; Dirac, A.M.; Ludwig, I.S.; Speijer, D.; Lindquist, S.; Vestman, E.-L.; Stax, M.J.; Geijtenbeek, T.B.H.; Pollakis, G.; Hernell, O.; et al. Bile Salt-Stimulated Lipase from Human Milk Binds DC-SIGN and Inhibits Human Immunodeficiency Virus Type 1 Transfer to CD4+ T Cells. Antimicrob. Agents Chemother. 2006, 50, 3367–3374. [Google Scholar] [CrossRef] [Green Version]
- Zhou, F.; Paz, H.A.; Sadri, M.; Cui, J.; Kachman, S.D.; Fernando, S.C.; Zempleni, J. Dietary bovine milk exosomes elicit changes in bacterial communities in C57BL/6 mice. Am. J. Physiol. Liver Physiol. 2019, 317, G618–G624. [Google Scholar] [CrossRef]
- Tong, L.; Hao, H.; Zhang, X.; Zhang, Z.; Lv, Y.; Zhang, L.; Yi, H. Oral Administration of Bovine Milk-Derived Extracellular Vesicles Alters the Gut Microbiota and Enhances Intestinal Immunity in Mice. Mol. Nutr. Food Res. 2020, 64, e1901251. [Google Scholar] [CrossRef]
- Shi, C.; Zhang, M.; Tong, M.; Yang, L.; Pang, L.; Chen, L.; Xu, G.; Chi, X.; Hong, Q.; Ni, Y.; et al. miR-148a is Associated with Obesity and Modulates Adipocyte Differentiation of Mesenchymal Stem Cells through Wnt Signaling. Sci. Rep. 2015, 5, 9930. [Google Scholar] [CrossRef] [Green Version]
- Behera, J.; Tyagi, N. Exosomes: Mediators of bone diseases, protection, and therapeutics potential. Oncoscience 2018, 5, 181–195. [Google Scholar] [CrossRef] [Green Version]
- Murata, T.; Takayama, K.; Katayama, S.; Urano, T.; Horie-Inoue, K.; Ikeda, K.; Takahashi, S.; Kawazu, C.; Hasegawa, A.; Ouchi, Y.; et al. miR-148a is an androgen-responsive microRNA that promotes LNCaP prostate cell growth by repressing its target CAND1 expression. Prostate Cancer Prostatic Dis. 2010, 13, 356–361. [Google Scholar] [CrossRef]
- Meng, F.; Henson, R.; Wehbe–Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 Regulates Expression of the PTEN Tumor Suppressor Gene in Human Hepatocellular Cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef] [Green Version]
- Feng, Y.-H.; Wu, C.-L.; Tsao, C.-J.; Chang, J.-G.; Lu, P.-J.; Yeh, K.-T.; Uen, Y.-H.; Lee, J.-C.; Shiau, A.-L. Deregulated expression of sprouty2 and microRNA-21 in human colon cancer: Correlation with the clinical stage of the disease. Cancer Boil. Ther. 2011, 11, 111–121. [Google Scholar] [CrossRef] [Green Version]
- Guo, S.L.; Peng, Z.; Yang, X.; Fan, K.J.; Ye, H.; Li, Z.H.; Wang, Y.; Xu, X.L.; Li, J.; Wang, Y.L.; et al. miR-148a Promoted Cell Proliferation by Targeting p27 in Gastric Cancer Cells. Int. J. Boil. Sci. 2011, 7, 567–574. [Google Scholar] [CrossRef]
- Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef]
- Betker, J.L.; Angle, B.M.; Graner, M.; Anchordoquy, T.J. The Potential of Exosomes From Cow Milk for Oral Delivery. J. Pharm. Sci. 2019, 108, 1496–1505. [Google Scholar] [CrossRef]
- Schiffelers, R.M.; Kooijmans, S.A.; Vader, P.; Van Dommelen, S.M.; Van Solinge, W.W. Exosome mimetics: A novel class of drug delivery systems. Int. J. Nanomed. 2012, 7, 1525–1541. [Google Scholar] [CrossRef] [Green Version]
- Murphy, D.E.; De Jong, O.G.; Brouwer, M.; Wood, M.J.; Lavieu, G.; Schiffelers, R.M.; Vader, P. Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp. Mol. Med. 2019, 51, 1–12. [Google Scholar] [CrossRef]
- Bamrungsap, S.; Zhao, Z.; Chen, T.; Wang, L.; Li, C.; Fu, T.; Tan, W. Nanotechnology in therapeutics: A focus on nanoparticles as a drug delivery system. Nanomedicine 2012, 7, 1253–1271. [Google Scholar] [CrossRef] [PubMed]
- Lim, E.-K.; Jang, E.; Lee, K.; Haam, S.; Huh, Y.-M. Delivery of Cancer Therapeutics Using Nanotechnology. Pharmaceutics 2013, 5, 294–317. [Google Scholar] [CrossRef]
- Dowdy, S.F. Overcoming cellular barriers for RNA therapeutics. Nat. Biotechnol. 2017, 35, 222–229. [Google Scholar] [CrossRef]
- Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2015, 371, 48–61. [Google Scholar] [CrossRef] [Green Version]
- Somiya, M.; Yoshioka, Y.; Ochiya, T. Biocompatibility of highly purified bovine milk-derived extracellular vesicles. J. Extracell. Vesicles 2018, 7, 1440132. [Google Scholar] [CrossRef] [Green Version]
- Agrawal, A.; Aqil, F.; Jeyabalan, J.; Spencer, W.A.; Beck, J.; Gachuki, B.W.; AlHakeem, S.; Oben, K.; Munagala, R.; Bondada, S.; et al. Milk-derived exosomes for oral delivery of paclitaxel. Nanomed. Nanotechnol. Boil. Med. 2017, 13, 1627–1636. [Google Scholar] [CrossRef]
- Aqil, F.; Kausar, H.; Agrawal, A.; Jeyabalan, J.; Kyakulaga, A.-H.; Munagala, R.; Gupta, R.C. Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp. Mol. Pathol. 2016, 101, 12–21. [Google Scholar] [CrossRef]
- Vashisht, M.; Rani, P.; Onteru, S.K.; Singh, D. Curcumin Encapsulated in Milk Exosomes Resists Human Digestion and Possesses Enhanced Intestinal Permeability in Vitro. Appl. Biochem. Biotechnol. 2017, 183, 993–1007. [Google Scholar] [CrossRef]
- Munagala, R.; Aqil, F.; Jeyabalan, J.; Agrawal, A.; Mudd, A.M.; Kyakulaga, A.H.; Singh, I.P.; Vadhanam, M.V.; Gupta, R.C. Exosomal formulation of anthocyanidins against multiple cancer types. Cancer Lett. 2017, 393, 94–102. [Google Scholar] [CrossRef] [Green Version]
- Aqil, F.; Jeyabalan, J.; Agrawalb, A.; Kyakulaga, A.-H.; Munagala, R.; Parker, L.; Gupta, R.C. Exosomal delivery of berry anthocyanidins for the management of ovarian cancer. Food Funct. 2017, 8, 4100–4107. [Google Scholar] [CrossRef]
- Kooijmans, S.A.; Schiffelers, R.M.; Zarovni, N.; Vago, R. Modulation of tissue tropism and biological activity of exosomes and other extracellular vesicles: New nanotools for cancer treatment. Pharmacol. Res. 2016, 111, 487–500. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.; Robbins, P.D. Immunosuppressive Exosomes: A New Approach for Treating Arthritis. Int. J. Rheumatol. 2012, 2012, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arntz, O.J.; Pieters, B.C.; Oliveira, M.C.; Broeren, M.; Bennink, M.B.; De Vries, M.; Van Lent, P.L.; Koenders, M.; Berg, W.B.V.D.; Van Der Kraan, P.; et al. Oral administration of bovine milk derived extracellular vesicles attenuates arthritis in two mouse models. Mol. Nutr. Food Res. 2015, 59, 1701–1712. [Google Scholar] [CrossRef]
- Lluis, A.; Depner, M.; Gaugler, B.; Saas, P.; Casaca, V.I.; Raedler, D.; Michel, S.; Tost, J.; Liu, J.; Genuneit, J.; et al. Increased regulatory T-cell numbers are associated with farm milk exposure and lower atopic sensitization and asthma in childhood. J. Allergy Clin. Immunol. 2014, 133, 551–559.e10. [Google Scholar] [CrossRef]
Milk Source | Main Proteins Implicated | Method of Isolation and Characterization | Reference |
---|---|---|---|
Bovine | Butyrophillin, Xanthine Oxidase, Adipophilin, Lactadherin | Differential centrifugation, ultracentrifugation, sucrose gradient, LC-MS/MS | [108] |
Bovine (S. aureus infected cows) | Butyrophillin, Xanthine dehydrogenase, Lactadherin, fatty acid synthase | Differential centrifugation, ultracentrifugation, sucrose gradient, LC-MS/MS | [130] |
Human | CD9, Annexin A5, Flotillin-1, CD83, CD81, Lactadherin, Syntenin, Rab, Ras-related proteins | Differential centrifugation, ultracentrifugation, sucrose gradient, LC-MS/MS | [123] |
Horse | CD81, CD63 receptors, Beta-Lactoglobulin, Lactadherin, Butyrophillin, Lactoferrin, Xanthine dehydrogenase | Differential centrifugation, ultracentrifugation, sucrose gradient, MALDI MS/MS | [107] |
Porcine | CD9, CD63, HSPs, Lactadherin, Butyrophillin, Adipophilin, Xanthine oxidase | Differential centrifugation, ultracentrifugation, sucrose gradient, LC-ESI-MS/MS | [106] |
Human | MHC II, CD81, MUC-1, HSPs, CD63, Butyrophillin, Lactadherin | Differential centrifugation, ultracentrifugation, sucrose gradient, LC-MS/MS | [42] |
Bovine | Butyrophillin, Xanthine Oxidase, Adipophilin, Lactadherin, Rab GTPases, integrins | Differential centrifugation, ultracentrifugation, sucrose gradient, LC-MS/MS | [66] |
Milk Source | Biomolecule (Number) | Implication | Method of Isolation | Reference |
---|---|---|---|---|
Human | miRNA (602) | Immunoregulatory, infant gut development | Differential centrifugation, ExoQuick exosome precipitation | [110] |
Bovine | miRNA (27) | Immune modulation | Differential centrifugation, ultracentrifugation, sucrose gradient | [132] |
Bovine | mRNA (19,320), miRNA (79) | Immune modulation | Differential centrifugation, ultracentrifugation | [102] |
Bovine (S. aureus infected cows) | miRNA (417) | Immunoregulation | Differential centrifugation, ultracentrifugation, sucrose gradient | [27] |
Porcine | mRNA (19,230) | Metabolism, signalling pathways | Differential centrifugation, ultracentrifugation, sucrose gradient | [106] |
Human | miRNA (330, 308) | Early infant development | Differential centrifugation, ExoQuick-TC | [118] |
Bovine | miRNA (69) | Signalling pathways | Differential centrifugation, ultrafiltration, ExoEasy Maxi Kit | [133] |
Bovine | miRNA (334) | Gene expression regulation | Differential centrifugation, ultracentrifugation, ultrafiltration | [125] |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sanwlani, R.; Fonseka, P.; Chitti, S.V.; Mathivanan, S. Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery. Proteomes 2020, 8, 11. https://doi.org/10.3390/proteomes8020011
Sanwlani R, Fonseka P, Chitti SV, Mathivanan S. Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery. Proteomes. 2020; 8(2):11. https://doi.org/10.3390/proteomes8020011
Chicago/Turabian StyleSanwlani, Rahul, Pamali Fonseka, Sai V. Chitti, and Suresh Mathivanan. 2020. "Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery" Proteomes 8, no. 2: 11. https://doi.org/10.3390/proteomes8020011
APA StyleSanwlani, R., Fonseka, P., Chitti, S. V., & Mathivanan, S. (2020). Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery. Proteomes, 8(2), 11. https://doi.org/10.3390/proteomes8020011