Next Article in Journal
Flying Together: Drosophila as a Tool to Understand the Genetics of Human Alcoholism
Next Article in Special Issue
The Role of Exosomes in Stemness and Neurodegenerative Diseases—Chemoresistant-Cancer Therapeutics and Phytochemicals
Previous Article in Journal
mRNA Post-Transcriptional Regulation by AU-Rich Element-Binding Proteins in Liver Inflammation and Cancer
Previous Article in Special Issue
Updated Understanding of Cancer as a Metabolic and Telomere-Driven Disease, and Proposal for Complex Personalized Treatment, a Hypothesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Milk Exosomes: Perspective Agents for Anticancer Drug Delivery

1
SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
2
Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(18), 6646; https://doi.org/10.3390/ijms21186646
Submission received: 10 August 2020 / Revised: 5 September 2020 / Accepted: 9 September 2020 / Published: 11 September 2020
(This article belongs to the Special Issue Exosomes in Cancer Diagnosis and Therapy)

Abstract

:
Exosomes are biological nanovesicles that participate in intercellular communication by transferring biologically active chemical compounds (proteins, microRNA, mRNA, DNA, and others). Due to their small size (diameter 40–100 nm) and high biological compatibility, exosomes are promising delivery tools in personalized therapy. Because artificial exosome synthesis methods are not developed yet, the urgent task is to develop an effective and safe way to obtain exosomes from natural sources. Milk is the only exosome-containing biological fluid that is commercially available. In this regard, milk exosomes are unique and promising candidates for new therapeutic approaches to treating various diseases, including cancer. The appearance of side effects during the use of cytotoxic and cytostatic agents is among the main problems in cancer chemotherapy. According to this, the targeted delivery of chemotherapeutic agents can be a potential solution to the toxic effect of chemotherapy. The ability of milk exosomes to carry out biologically active substances to the cell makes them promising tools for oral delivery of chemotherapeutic agents. This review is devoted to the methods of milk exosome isolation, their biological components, and prospects for their use in cancer treatment.

1. Introduction

Exosomes are natural extracellular nanovesicles that participate in intercellular communication by carrying biologically active substances like proteins, microRNA, mRNA, DNA, and other molecules [1,2]. The carried compounds can be transported both inside and on the surface of exosomes [3]. Due to their small size (diameter 40–100 nm), exosomes are promising delivery tools in personalized therapy [4]. Because artificial exosome synthesis methods are not developed yet, the urgent task is to effectively and safely obtain exosomes in mass quantities from natural sources. Tumor cell cultures can hardly be considered as a promising source of vesicles for pharmacology applications. The exosome content is shown in various biological fluids: blood plasma, urine, saliva, milk, amniotic fluid, ascites, cerebrospinal fluid, and others [2]. Among them, milk is the only biological liquid containing exosomes that is available on an industrial scale.
Human milk exosomes were first described in 2007. To date, exosomes have been isolated (chronologically) from human [5], bovine [6], porcine [7], wallaby [8], camel [9], rat [10], horse [11], panda [12], yak [13], sheep [14], and goat milk [15]. Databases provide more than 100 articles on exosomes and other milk vesicles (more than half of them are on human and cow milk vesicles). Large volumes of milk can be obtained from a cow and relatively large from horse, sheep, goat, and camel. Due to possible prion content, bovine milk should not be considered a source of protein-containing structures [16].
Milk exosomes are promising candidates in developing new therapeutic approaches to the treatment of various diseases, including cancer. However, currently, there is only incomplete data in the literature on the use of milk vesicles, particularly exosomes, to deliver biologically active molecules to cells [17]. In the application of milk exosomes for cancer treatment, two directions can be highlighted: the delivery of antitumor drugs [18] and therapeutic nucleic acids [19,20]. Various side effects occurring during the use of cytotoxic and cytostatic agents are unresolved problems of cancer chemotherapy [21]. Cells with a high mitotic index are most susceptible to the action of cytostatic agents; thus, the use of cytostatic drugs leads to the death of not only the tumor cells but also cells of the bone marrow, skin, hair, epithelium of the gastrointestinal tract, etc. [22]. Targeted chemotherapeutic agent delivery can be a potential solution to the occurrence of toxic effects in chemotherapy [4,23].
Because exosomes contain mRNA, microRNA, and proteins that may harm cells, some experts disagree that exosomes (including milk exosomes) can be used as delivery vehicles. Analysis of literature data shows that many papers devoted to the protein and nucleic components of exosomes may have overestimated the number of proteins and/or nucleic acid molecules in exosomes due to contamination of the preparations with co-isolating molecules [24,25]. Therefore, it is essential to analyze highly purified exosome preparations to identify biopolymers that are intrinsic components of exosomes and the proteins and nucleic acids that co-isolate with exosomes. Exosomes can be used as drug delivery systems, carrying the drugs of therapeutic nucleic acids on the surface or after “loading” of these components using electroporation, sonication, or ligands on the surface of exosomes [26]. Another method is the fusion of exosomes with liposomes, loaded with necessary contents to form “chimeric” exosomes [27]. We propose that the use of highly purified exosomes as drug delivery vehicles is critical to avoid unwanted side effects. This review is devoted to the methods of milk exosome isolation, analysis of their biological components, and prospects for their use in cancer treatment.

2. Isolation of Milk Exosomes

Various physical, physicochemical, and immunological approaches are used to isolate exosomes from milk. General issues concerning the isolation and characterization of exosomes are described in the guidelines of the International Society for Extracellular Vesicles [28,29]. Because the volume of milk obtained from single humans or animals may vary from milliliters to liters, the general protocol [1] of exosome isolation from milk samples usually starts with a series of centrifugations [6]: first, several sequential centrifugations at low speed of 600–1000× g to defat the milk and precipitate the cells, then 10,000–16,000× g to precipitate milk proteins, followed by one or several ultracentrifugations at 100,000–200,000× g [30] to isolate the exosomes. Some protocols include the stage of ultracentrifugation in the sucrose density gradient [31].
Patented and commercially available sets of reagents using volume-excluding polymers are also used: for example, ExoQuick (System Biosciences, Palo Alto, CA, USA)—contains PEG 8000 kDa; Total Exosome Isolation (Thermo Fisher Scientific, Waltham, MA, USA)—includes dextrans, polyethylene glycol, polyvinyl with a molecular weight above 1000 kDa, and their combinations with other methods [32]. Different exosome isolation methods include filtration through a semipermeable membrane and isolation on immunomagnetic particles [4].
One- or two-time centrifugation of milk at low speeds before subsequent optional storage of milk plasma preliminary to exosome isolation is considered fundamental, as described in many protocols, for example, [30]. Because the composition of nucleic acids, proteins [33], and lipids [34] of milk exosomes and milk fat globule membrane (MFGM) in various articles often coincides, it cannot be excluded that storage of nondefatted milk samples leads to contamination of exosome preparations with MFGM. Refrigeration of nondefatted milk samples may lead to exosomes sticking to MFGM, resulting in further MFGM contamination of the exosome samples obtained.
Exosome preparations isolated by centrifugation and ultracentrifugation from blood plasma, urine, milk, lacrimal, and cell culture fluid usually contain “nonvesicles” with 20–100 nm diameter. These “nonvesicles” are morphologically attributed to intermediate- and low-density lipoproteins (20–40 nm) and very low-density (40–100 nm) lipoproteins [35]. These have a different structure according to transmission electron microscopy and should not be confused with exosomes.
The possibility of exosome isolation using gel filtration after or instead of ultracentrifugation [36] has been shown for human [37], bovine [38], and horse milk. Because the gel filtration allows us to effectively separate sample volumes that do not exceed 1–5% of the column volume, this method is not suitable for isolating exosomes directly from large amounts of milk. Gel filtration allows purifying the sample from co-isolating proteins in preparations obtained after ultrafiltration and/or ultracentrifugation. Our data suggest that sequential centrifugations and ultracentrifugations followed by the ultrafiltration throw 0.22 µm filter are not enough to get purified exosomes [39]. Moreover, gel filtration of sediments obtained by ultracentrifugation results in exosome preparations without admixtures of co-isolating proteins [11].

3. Bioactive Compounds of Milk Exosomes

The biochemical components of milk exosomes—proteins, lipids, and nucleic acids—can significantly affect therapeutic molecule delivery. In this regard, a detailed analysis of the content of these molecules in milk exosomes, also called “exosomics” (by analogy with genomics, proteomics, and other -omics technologies), is required [40].

3.1. Milk Exosome Proteins

The articles devoted to proteomic analysis of milk exosomes using modern highly sensitive methods describe dozens, hundreds, and even thousands of proteins and peptides, such as 115 [41], 571 [42], 2107 [43] or 2698 [44] individual proteins, and isoforms and 719 peptides [45]. According to the critical article [25], these numbers are greatly exaggerated, most likely, due to the attribution of co-isolating milk proteins with exosomes [39,46].
The milk exosomes contain several proteins that participate in their formation: Testilin controls membrane fusion [5], Rab GTPase interacts with cytoskeleton proteins [47], and Alix and Tsg101 are involved in endocytosis [48]. Moreover, milk exosomes contain proteins that determine their biological functions, namely, transport of microRNA and adhesion to the target cells (tetraspanins CD9, CD63, CD81) [49,50]. Functional activity of milk exosomal enzymes—fatty acid synthase, xanthine dehydrogenase, proteases, ADP-ribosylation factor—is not yet evident [39]. Milk exosomes also contain cytoskeleton proteins—actin, tubulin, cofilin, heat shock proteins, and molecules—involved in signal transduction, for example, in the Wnt signaling pathway [51,52]. Nuclear, mitochondrial, reticulum, and Golgi proteins cannot be an intrinsic part of exosomes as they are not transported to endosomes [53]. In any case, because the exosomes originate from an independent cellular compartment, their protein composition is not accidental.
Previously, we proposed an original method for isolating exosomes, which allows us to obtain preparations that practically do not contain co-isolating proteins from two or more liters of human of horse milk [11,24]. According to these results, horse milk exosomes contain mainly actin, butyrophilin, β-lactoglobulin, lactadherin, lactoferrin, and xanthine dehydrogenase, as well as numerous peptides (see Table 1).
These results correspond to the literature data obtained using highly sensitive proteomic methods [39], according to which butyrophilin, lactadhedrin, and xanthine dehydrogenase are the specific markers of milk exosomes. Because α-, β-, and κ-caseins and ribosomal proteins cannot be present in exosome preparations according to incompatible secretion mechanisms [40,59], these proteins are not intrinsic components and may be considered as “negative markers” of milk exosomes (fractions containing these protein markers are not exosomes or contain co-isolated protein impurities).

3.2. Nucleic Acids of Milk Exosomes

Studies of the past decade have shown the content of various noncoding RNA—microRNA, long noncoding RNA, circular RNA [60]—in bovine [61], human [62], panda [12], porcine [7], and rat [63] milk. Exosomal RNAs are stable at low intestinal pH values and in the presence of RNases [61,64]. Milk mRNAs are mainly concentrated in exosomes, while microRNAs are concentrated in exosomes and the supernatant [65].
The microRNA contents of milk exosomes are analyzed by high throughput sequencing (NGS) and microarray technology. Analysis of global expression profiles using microarrays revealed 79 different microRNAs (miRNAs) in the exosomal fraction and 91 in the supernatant after ultracentrifugation of bovine milk, and 39 miRNAs were common for both fractions. Further studies have shown that the expression level of these microRNAs is significantly higher in the exosomal portion compared to the supernatant [65]. Some 491 miRNAs were described in porcine milk exosomes, including 176 known miRNAs and 315 new mature miRNAs. Analysis of the gene ontology of these microRNAs showed that most of them are targeting genes related to transcriptional, immune, and metabolic processes [66]. In a study of the microRNA profile in exosomes of human milk during type I diabetes mellitus, 631 exosomal microRNAs were identified, including immune-related ones such as hsa-let-7c, hsa-miR-21, hsa-miR-34a, hsa-miR- 146b, and hsa-miR-200b. Differential expression was described for nine miRNAs in healthy and diabetic groups [67]. MircroRNAs, highly represented in milk exosomes, are reviewed in Table 2.
Different microRNA of milk and milk exosomes usually have several targets and are shown to be important in processes of fetal development, cell proliferation, pregnancy, immune system development, inflammation, sugar metabolism, insulin resistance [1,71], and many others (Table 3).
Literature data provide information on the content of 16,304 different mRNAs in porcine milk exosomes [42] and up to 19,230 mRNAs in bovine milk exosomes [65]. As with thousands of proteins, it is difficult to imagine how these thousands of mRNAs can fit in a 40–100 nm vesicle [25]. Interestingly, 18S and 28S ribosomal RNAs are practically absent in the milk-derived exosomes of human [49] and porcine milk [7], which corresponds to the current knowledge biogenesis of these vesicles.
We assume that one should be critical of information about the content of thousands of microRNA molecules and, especially, mRNA in milk exosomes. The mechanism by which these nucleic acids can co-isolate with vesicles is not as apparent as for proteins. The potential interaction of nucleic acids in milk exosomes (especially the anti-inflammatory effect and weakening of the immune response [1]) should also be taken into account when planning experiments on the delivery of therapeutic nucleic acids to cells.

3.3. Lipids of Milk Exosomes

Exosomes are vesicles rounded by the lipid bilayer containing proteins directed to the extracellular space. In this regard, the delivery of pharmacologically significant compounds is possible both inside and outside exosomes, both for hydrophilic (bound to surface proteins) and hydrophobic (as part of the lipid bilayer) molecules. The exosomal membrane is known to contain cholesterol, phosphatidylcholine, phosphatidylinositol, sphingomyelin, and ceramides; most phosphatidylcholine and sphingolipids has been shown to be located in the outer layer of the exosomal membrane [77]. The thickness of the lipid bilayer is at least 5 nm, so the exosome’s minimum diameter cannot be less than 40 nm. Depending on the source and the methods used, the composition of exosome lipids in milk differs from one source to another. A detailed overview of exosome lipids is given in [77,78].
The problem of exosome preparation’s contamination with lipid droplets, lipoproteins, and other particles [78] during their isolation by centrifugation and ultracentrifugation limits the study of intrinsic exosomal lipids. Exosomes isolated from cell cultures contain arachidonic acid, prostaglandins, enzymes of fatty acid metabolism [55], and leukotrienes [79]. This data indicates the need for further study of lipids in highly purified exosome preparations.
Liposomes have been widely used as targeted delivery vehicles for over 40 years. Liposome membranes can be modified to improve the targeting properties with monoclonal antibodies, aptamers, proteins, and small molecules [26]. A promising way of changing the membrane of natural exosomes is its fusion to liposomes with artificially functionalized lipids [4]. Polymer nanoparticles might have higher stability compared to liposomes, but their biocompatibility and safety in the case of long-term administration remain an unresolved problem [80]. Natural lipid components of milk exosomes are more compatible and less toxic than artificial liposomes for use as drug delivery vehicles [81].
To increase the efficiency of drug delivery by milk exosomes, a more intensive study of the lipid composition of exosomes, proteins that are located in the lipid membrane, is required. Modification of exosomal surface for targeted delivery and prolonged half-life in the body is an essential task for biopharmacology and biomedicine [82].

4. Milk Exosomes for Drug Delivery in Cancer Therapy

The use of antitumor drugs on the surface of natural biologically active structures, such as proteins, significantly increases the therapy’s biological availability and effectiveness [83]. Recently, much attention has been paid to the ability of exosomes to overcome physiological barriers, including the blood–brain barrier [84,85]. In this regard, many laboratories have attempted to use exosomes isolated from various cell lines for targeted drug delivery [86,87]. Despite the remarkable results shown, there are significant challenges in the development of exosomes as therapeutic products. The collection of exosomes from cell lines and/or patient samples is not compatible with commercial pharmaceutical production, and the protein component of exosomes is likely to trigger immune responses when administered systemically [85,88]. These can be overcome by using easily accessible mass sources of exosomes while solving immune responses. Studies have revealed that exosomes can be obtained from bovine milk in scalable quantities [89]; at the same time, the introduction of milk exosomes did not cause systemic toxicity and anaphylactic effect in mouse models [90]. Nonloaded camel milk exosomes contributed to a significant reduction in the progression of breast tumors, as evidenced by increased markers of apoptotic processes as well as oxidative stress reduction and downregulation of several genes associated with inflammatory processes and induction of the immune response [91]. Because the widespread consumption of milk and dairy products probably contributed to the development of human immune tolerance to milk proteins, milk exosomes can be used as delivery systems for oral drug administration [92].
In 2016, the targeting of milk exosomes to a malignant tumor was shown for the first time. Drug-loaded exosomes showed higher efficacy than free drugs in the treatment of cell cultures and xenografts of lung tumors in vivo. Milk exosomes showed interspecific tolerance without any adverse immune and inflammatory responses. Thus, the versatility of milk exosomes concerning the carrying load and the ability to reach the targeted tumor was shown [18]. In the study of milk exosomes for the delivery of the chemotherapeutic agent paclitaxel, a slight increase in the size of loaded exosomes was demonstrated, explained by the authors as partial inclusion of paclitaxel in the lipid bilayer of the membrane. At the same time, exosomes containing the drug were stable in simulated gastrointestinal conditions, highlighting their suitability for oral drug delivery [17].
Milk exosomes are used to deliver curcumin as a potential antitumor agent [93]. Exosomes significantly improved stability, solubility, and bioavailability of curcumin in adverse conditions of the digestive tract compared to free curcumin [94]. However, in a later study, it was demonstrated that curcumin loaded into exosomes derived from intestinal epithelial cells was much more effectively absorbed by colorectal adenocarcinoma cells than curcumin in milk exosomes [95]. Celastrol, a plant-derived triterpenoid, has demonstrated anti-inflammatory, antiproliferative, and antitumor activity [96,97]. However, the use of this drug is highly restricted due to its low bioavailability and toxicity. Milk exosomes, loaded with celastrol, showed high antitumor activity compared to free celastrol on the xenograft model of nonsmall cell lung cancer. Simultaneously, celastrol delivered via milk exosomes did not show gross systemic toxicity [98]. Loading preparations based on berry anthocyanins into milk exosomes [99] contributed to a significant increase in the antiproliferative activity of the drug in the experiment with cisplatin-resistant ovarian cancer cell lines [100], which also emphasizes the prospects of exosomal encapsulation when using various antitumor agents.
One of the problems of milk exosome application for targeted drug delivery is the lack of specificity to recipient cells. It has been shown that milk exosomes are absorbed from the gut as intact particles that can be modified by ligands to promote retention in target tissues [101]. Like liposome-based drug delivery systems, milk exosome-based vectors can be equipped with particular ligands to bind tumor-specific receptors [102]. The CD44 receptor, with specific ligand hyaluronan, is often overexpressed on the surface of various types of cancer cells [103]. Equipping the lipid membrane of milk exosomes with hyaluronan molecules enabled targeted delivery of the cytostatic agent doxorubicin to cells expressing CD44 [102]. The addition of folic acid as a tumor receptor ligand to milk exosomes loaded with a chemotherapeutic agent significantly enhanced tumor cell growth inhibition in a mouse xenograft model [18]. Bovine milk exosomes in vitro activated CD69 (an early marker of lymphocyte proliferation) on the normal killer (NK) cells. The stimulation of γ-interferon (IFN) production by NK-cells and T-lymphocytes after co-activation with milk exosomes and interleukins 2 and 12 indicates the possible undesirable enhancement of the inflammatory process [104].
Among current trials registered on clinicaltrials.gov, none are using milk exosomes as drug delivery systems. Nevertheless, studies demonstrate that the use of milk exosomes for the delivery of antitumor drugs contributes to a significant increase in their effectiveness and reduces the toxicity of the therapy, which opens up excellent prospects for the application of milk exosomes for oral delivery of therapeutic agents [18]. Molecules delivered with milk exosomes are reviewed in Table 4.

5. Biological Activity of Milk Exosome Nucleic Acids and Delivery of Nucleic Acids to Cancer Cells

Cell lines of acute leukemia and colon cancer capture breast milk exosomes and their RNA contents. MicroRNAs from both the fat layer and the supernatant were detected in these cells. An increase in the expression level of miR-148a-3p and a decrease in the DNA methyltransferase I (DNMT1) gene, which is its direct target, were also detected [106]. These facts suggest that RNAs contained in milk exosomes can directly or indirectly affect the growth and development of cancer cells. Men who consumed more than three daily norms of high-fat bovine milk showed an increased risk of prostate cancer [107]. Milk exosomes supplemented to prostate cancer cell cultures increased their proliferative activity by 30% [108]. These effects are most likely related to the microRNA contents of human [109], porcine [66], and bovine [110] milk that can significantly reduce the expression level of the TP53 gene, with negative regulation one of the typical events in carcinogenesis. MicroRNA, presented in human milk exosomes, may control up to 9074 genes [109,111].
The main bovine milk microRNA miR-21 was found to target p57Kip2, a factor that inhibits cyclin-dependent kinases in prostate cancer cells [112]. Elevated levels of miR-21 were found in the serum and tumor tissue of breast cancer patients [113,114]. P57kip2 knockdown enhances the proliferative phenotype induced by tumor-associated mutant variants of phosphoinositide 3-kinase. It releases mammary epithelial acinus from cell arrest during morphogenesis [115], suggesting a potential effect of mammary miR-21 on tumor growth processes. Long noncoding RNA of porcine milk exosomes may interact with miRNA related to cell proliferation, and circular RNA may target miRNA associated with intestinal cells [60]. A recent paper showed that miRNAs from bovine and porcine milk were adsorbed in piglet intestine and transported to the blood serum with different efficiencies [92].
Exosomes stimulate the proliferation of intestinal epithelium cells of healthy but not tumor cultures. Incubation of healthy cells with milk exosomes results in a decrease of the phosphatase and tensin homolog (PTEN) expression level, which is the main target of miR-148a, the content of which has also been shown for milk exosomes [65,74]. In miR-148a knockout cells, milk exosomes suppress the proliferation and expression of DNMT1 [116]. Thus, studies have shown the content of many different microRNAs in milk exosomes and their bioavailability and ability to modify the expression of multiple genes, including those associated with the occurrence and growth of malignant tumors.
The ability of exosomes to transfer mRNA and microRNA between cells and subsequently mediate changes in target gene expression in recipient cells also highlights their potential for delivering exogenous small interfering RNAs. Small interfering RNAs (siRNAs) are a potential generation of new therapeutics. With increasing knowledge on the endogenous RNA interference, the possibility of using siRNA as nucleic acid-based drugs for many diseases, including different types of cancer, is growing [117]. Despite the tremendous therapeutic potential, siRNAs are hindered by the lack of effective ways to deliver them to the cell. Chemical modification of small interfering RNAs for their delivery was unsuccessful and, in some cases, led to the loss of their biological activity [118,119] and the occurrence of toxic effects [120]. Polymer nanoparticles, lipids and liposomes, peptides, and synthetic nanocarriers were considered alternative options for delivering siRNA to cells; the most developed delivery system is based on liposomes [121]. However, these methods never solved the problem of nonspecific targeting and immune response. Bovine milk exosomes can deliver endogenous RNA load to recipient cells, and it remains stable, resisting degradation [105]. The dose-dependent antiproliferative activity of exosomes loaded with siRNAs interfering with the mutant allele of the KRAS gene in cell lines and mouse xenografts of the lung cancer were demonstrated [20].

6. Conclusions

The development of biocompatible methods of delivering therapeutically significant drugs in vitro and in vivo is essential for molecular pharmacology. Exosomes of horse, goat, and sheep milk are free of prion proteins [16] and have excellent prospects for practical application. Still, some problems limiting the use of milk exosomes in therapy remain unresolved: insufficient data on both proteins and nucleic acids that are an intrinsic part of exosomes and which are co-isolated during purification; the specificity of milk exosomes to various cells and tissues; and the development of methods for modifying the surface of milk exosomes for target delivery.
Delivery by artificially produced liposomes is often compared with natural exosomes, and various reasons are given pro et contra use of natural vesicles. The main advantage of milk exosomes for drug delivery compared to liposomes and other artificial nanoparticles is higher biocompatibility, namely, lower immunogenicity and cytotoxicity [122], due to the chemical composition of exosomes similar to cell membranes [80]. Milk exosomes can deliver both hydrophilic and hydrophobic molecules; the isolation from milk is scalable and cheaper compared with isolation from blood plasma or culture fluid.
Biologically active molecules carried by milk exosomes remain stable even in the digestive tract’s harsh conditions, making it possible to use milk exosomes for oral drug delivery [18,101]. The described diversity of various miRNAs in the composition of milk exosomes and their influence on the implementation of genetic information in recipient cells opens up a vast potential for exosomes in gene therapy of diseases, especially oncological ones. The lipid composition of milk exosomes, which plays an essential role in the delivery of drugs and biologically active molecules, is also insufficiently studied. The facts above indicate the relevance for further study of the biochemical composition of milk exosomes.

Author Contributions

Conceptualization, S.S.; writing—original draft preparation, S.S., and A.K.; writing—review and editing, S.S. and G.N.; supervision, G.N.; project administration, S.S.; funding acquisition, S.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the RSF, grant number 18-74-10055, to Sergey Sedykh and the Russian state-funded budget project of ICBFM SB RAS # AAAA-A17-117020210023-1 to Georgy Nevinsky.

Acknowledgments

The authors thank Elena V. Ryabchikova and RAS Valentin V. Vlassov for inspirational ideas and discussions.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

DNMTDNA methyltransferase
IFNinterferon
ILinterleukin
MFGMmilk fat globule membrane
NKnormal killer
PTENphosphatase and tensin homolog
miRNAmicroRNA
siRNAsmall interfering RNA

References

  1. Cintio, M.; Polacchini, G.; Scarsella, E.; Montanari, T.; Stefanon, B.; Colitti, M. MicroRNA Milk Exosomes: From Cellular Regulator to Genomic Marker. Animals 2020, 10, 1126. [Google Scholar] [CrossRef] [PubMed]
  2. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Annu. Rev. Cell Dev. Biol 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
  3. Sawada, S.I.; Sato, Y.T.; Kawasaki, R.; Yasuoka, J.I.; Mizuta, R.; Sasaki, Y.; Akiyoshi, K. Nanogel hybrid assembly for exosome intracellular delivery: Effects on endocytosis and fusion by exosome surface polymer engineering. Biomater. Sci. 2020, 8, 619–630. [Google Scholar] [CrossRef] [PubMed]
  4. Bunggulawa, E.J.; Wang, W.; Yin, T.; Wang, N.; Durkan, C.; Wang, Y.; Wang, G. Recent advancements in the use of exosomes as drug delivery systems. J. Nanobiotechnol. 2018, 16, 81. [Google Scholar] [CrossRef] [Green Version]
  5. 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]
  6. Yamada, T.; Inoshima, Y.; Matsuda, T.; Ishiguro, N. Comparison of methods for isolating exosomes from bovine milk. J. Vet. Med. Sci. 2012, 74, 1523–1525. [Google Scholar] [CrossRef] [Green Version]
  7. Gu, Y.; Li, M.; Wang, T.; Liang, Y.; Zhong, Z.; Wang, X.; Zhou, Q.; Chen, L.; Lang, Q.; He, Z.; et al. Lactation-related microRNA expression profiles of porcine breast milk exosomes. PLoS ONE 2012, 7. [Google Scholar] [CrossRef]
  8. Modepalli, V.; Kumar, A.; Hinds, L.A.; Sharp, J.A.; Nicholas, K.R.; Lefevre, C. Differential temporal expression of milk miRNA during the lactation cycle of the marsupial tammar wallaby (Macropus eugenii). BMC Genom. 2014, 15, 1012. [Google Scholar] [CrossRef] [Green Version]
  9. Yassin, A.M.; Abdel Hamid, M.I.; Farid, O.A.; Amer, H.; Warda, M. Dromedary milk exosomes as mammary transcriptome nano-vehicle: Their isolation, vesicular and phospholipidomic characterizations. J. Adv. Res. 2016, 7, 749–756. [Google Scholar] [CrossRef] [Green Version]
  10. Hock, A.; Miyake, H.; Li, B.; Lee, C.; Ermini, L.; Koike, Y.; Chen, Y.; Määttänen, P.; Zani, A.; Pierro, A. Breast milk-derived exosomes promote intestinal epithelial cell growth. J. Pediatr. Surg. 2017, 52, 755–759. [Google Scholar] [CrossRef]
  11. 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]
  12. Ma, J.; Wang, C.; Long, K.; Zhang, H.; Zhang, J.; Jin, L.; Tang, Q.; Jiang, A.; Wang, X.; Tian, S.; et al. Exosomal microRNAs in giant panda (Ailuropoda melanoleuca) breast milk: Potential maternal regulators for the development of newborn cubs. Sci. Rep. 2017, 7, 1–11. [Google Scholar] [CrossRef] [PubMed]
  13. Gao, H.N.; Guo, H.Y.; Zhang, H.; Xie, X.L.; Wen, P.C.; Ren, F.Z. Yak milk-derived exosomes promote proliferation of intestinal epithelial cells in hypoxic environment. J. Dairy Sci. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Quan, S.; Nan, X.; Wang, K.; Jiang, L.; Yao, J.; Xiong, B. Characterization of sheep milk extracellular vesicle-miRNA by sequencing and comparison with cow milk. Animals 2020, 10, 331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. González, M.I.; Martín-Duque, P.; Desco, M.; Salinas, B. Radioactive Labeling of Milk-Derived Exosomes with 99mTc and In Vivo Tracking by SPECT Imaging. Nanomaterials 2020, 10, 1062. [Google Scholar] [CrossRef] [PubMed]
  16. Franscini, N.; El Gedaily, A.; Matthey, U.; Franitza, S.; Sy, M.S.; Bürkle, A.; Groschup, M.; Braun, U.; Zahn, R. Prion protein in milk. PLoS ONE 2006, 1, e71. [Google Scholar] [CrossRef]
  17. Agrawal, A.K.; Aqil, F.; Jeyabalan, J.; Spencer, W.A.; Beck, J.; Gachuki, B.W.; Alhakeem, S.S.; Oben, K.; Munagala, R.; Bondada, S.; et al. Milk-derived exosomes for oral delivery of paclitaxel. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1627–1636. [Google Scholar] [CrossRef]
  18. Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016, 371, 48–61. [Google Scholar] [CrossRef] [Green Version]
  19. Matsuda, A.; Patel, T. Milk-derived Extracellular Vesicles for Therapeutic Delivery of Small Interfering RNAs. In Methods in Molecular Biology; Humana Press: New York, NY, USA, 2018; Volume 1740, pp. 187–197. ISBN 9781493976522. [Google Scholar]
  20. Aqil, F.; Munagala, R.; Jeyabalan, J.; Agrawal, A.K.; Kyakulaga, A.H.; Wilcher, S.A.; Gupta, R.C. Milk exosomes—Natural nanoparticles for siRNA delivery. Cancer Lett. 2019, 449, 186–195. [Google Scholar] [CrossRef]
  21. Wheate, N.J.; Walker, S.; Craig, G.E.; Oun, R. The status of platinum anticancer drugs in the clinic and in clinical trials. Dalt. Trans. 2010, 39, 8113. [Google Scholar] [CrossRef] [Green Version]
  22. Schirrmacher, V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (Review). Int. J. Oncol. 2019, 54, 407–419. [Google Scholar] [CrossRef]
  23. Saari, H.; Lázaro-Ibáñez, E.; Viitala, T.; Vuorimaa-Laukkanen, E.; Siljander, P.; Yliperttula, M. Microvesicle- and exosome-mediated drug delivery enhances the cytotoxicity of Paclitaxel in autologous prostate cancer cells. J. Control. Release 2015, 220, 727–737. [Google Scholar] [CrossRef] [Green Version]
  24. Burkova, E.E.; Grigor’eva, A.E.; Bulgakov, D.V.; Dmitrenok, P.S.; Vlassov, V.V.; Ryabchikova, E.I.; Sedykh, S.E.; Nevinsky, G.A. Extra Purified Exosomes from Human Placenta Contain an Unpredictable Small Number of Different Major Proteins. Int. J. Mol. Sci. 2019, 20, 2434. [Google Scholar] [CrossRef] [Green Version]
  25. Sverdlov, E.D. Amedeo Avogadro’s cry: What is 1 µg of exosomes? BioEssays 2012, 34, 873–875. [Google Scholar] [CrossRef] [PubMed]
  26. Antimisiaris, S.G.; Mourtas, S.; Marazioti, A. Exosomes and Exosome-Inspired Vesicles for Targeted Drug Delivery. Pharmaceutics 2018, 4, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Sato, Y.T.; Umezaki, K.; Sawada, S.; Mukai, S.A.; Sasaki, Y.; Harada, N.; Shiku, H.; Akiyoshi, K. Engineering hybrid exosomes by membrane fusion with liposomes. Sci. Rep. 2016, 6, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Lötvall, J.; Hill, A.F.; Hochberg, F.; Buzás, E.I.; Di Vizio, D.; Gardiner, C.; Gho, Y.S.; Kurochkin, I.V.; Mathivanan, S.; Quesenberry, P.; et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles 2014, 1, 1–6. [Google Scholar] [CrossRef] [PubMed]
  29. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2019, 8. [Google Scholar] [CrossRef] [Green Version]
  30. Zonneveld, M.I.; Brisson, A.R.; van Herwijnen, M.J.C.; Tan, S.; van de Lest, C.H.A.; Redegeld, F.A.; Garssen, J.; Wauben, M.H.M.; Nolte-’t Hoen, E.N.M. Recovery of extracellular vesicles from human breast milk is influenced by sample collection and vesicle isolation procedures. J. Extracell. Vesicles 2014, 3, 24215. [Google Scholar] [CrossRef] [Green Version]
  31. Yamauchi, M.; Shimizu, K.; Rahman, M.; Ishikawa, H.; Takase, H.; Ugawa, S.; Okada, A.; Inoshima, Y. Efficient method for isolation of exosomes from raw bovine milk. Drug Dev. Ind. Pharm. 2019, 45, 359–364. [Google Scholar] [CrossRef]
  32. Rider, M.A.; Hurwitz, S.N.; Meckes, D.G. ExtraPEG: A Polyethylene Glycol-Based Method for Enrichment of Extracellular Vesicles. Sci. Rep. 2016, 6, 23978. [Google Scholar] [CrossRef] [PubMed]
  33. Yang, M.; Cong, M.; Peng, X.; Wu, J.; Wu, R.; Liu, B.; Ye, W.; Yue, X. Quantitative proteomic analysis of milk fat globule membrane (MFGM) proteins in human and bovine colostrum and mature milk samples through iTRAQ labeling. Food Funct. 2016, 7, 2438–2450. [Google Scholar] [CrossRef] [PubMed]
  34. Arranz, E.; Corredig, M. Invited review: Milk phospholipid vesicles, their colloidal properties, and potential as delivery vehicles for bioactive molecules. J. Dairy Sci. 2017, 100, 4213–4222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Grigor’eva, A.E.; Dyrkheeva, N.S.; Bryzgunova, O.E.; Tamkovich, S.N.; Chelobanov, B.P.; Ryabchikova, E.I. Contamination of exosome preparations, isolated from biological fluids. Biomed. Khim. 2017, 63, 91–96. [Google Scholar] [CrossRef] [Green Version]
  36. Blans, K.; Hansen, M.S.; Sørensen, L.V.; Hvam, M.L.; Howard, K.A.; Möller, A.; Wiking, L.; Larsen, L.B.; Rasmussen, J.T. Pellet-free isolation of human and bovine milk extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles 2017, 6, 1294340. [Google Scholar] [CrossRef]
  37. Vaswani, K.; Mitchell, M.D.; Holland, O.J.; Qin Koh, Y.; Hill, R.J.; Harb, T.; Davies, P.S.W.; Peiris, H. A Method for the Isolation of Exosomes from Human and Bovine Milk. J. Nutr. Metab. 2019, 2019, 1–6. [Google Scholar] [CrossRef]
  38. Vaswani, K.; Koh, Y.Q.; Almughlliq, F.B.; Peiris, H.N.; Mitchell, M.D. A method for the isolation and enrichment of purified bovine milk exosomes. Reprod. Biol. 2017, 17, 341–348. [Google Scholar] [CrossRef]
  39. Sedykh, S.E.; Burkova, E.E.; Purvinsh, L.V.; Klemeshova, D.A.; Ryabchikova, E.I.; Nevinsky, G.A. Milk Exosomes: Isolation, Biochemistry, Morphology, and Perspectives of Use. In Extracellular Vesicles and Their Importance in Human Health; IntechOpen: Rijeka, Croatia, 2020. [Google Scholar]
  40. de la Torre Gomez, C.; Goreham, R.V.; Bech Serra, J.J.; Nann, T.; Kussmann, M. “Exosomics”—A Review of Biophysics, Biology and Biochemistry of Exosomes With a Focus on Human Breast Milk. Front. Genet. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  41. 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]
  42. Chen, T.; Xi, Q.-Y.; Sun, J.-J.; Ye, R.-S.; Cheng, X.; Sun, R.-P.; Wang, S.-B.; Shu, G.; Wang, L.-N.; Zhu, X.-T.; et al. Revelation of mRNAs and proteins in porcine milk exosomes by transcriptomic and proteomic analysis. BMC Vet. Res. 2017, 13, 101. [Google Scholar] [CrossRef]
  43. Reinhardt, T.A.; Lippolis, J.D.; Nonnecke, B.J.; Sacco, R.E. Bovine milk exosome proteome. J. Proteom. 2012, 75, 1486–1492. [Google Scholar] [CrossRef] [PubMed]
  44. van Herwijnen, M.J.C.; Zonneveld, M.I.; Goerdayal, S.; Nolte’t Hoen, E.N.M.; Garssen, J.; Stahl, B.; Maarten Altelaar, A.F.; Redegeld, F.A.; Wauben, M.H.M. 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]
  45. Wang, X.; Yan, X.; Zhang, L.; Cai, J.; Zhou, Y.; Liu, H.; Hu, Y.; Chen, W.; Xu, S.; Liu, P.; et al. Identification and Peptidomic Profiling of Exosomes in Preterm Human Milk: Insights Into Necrotizing Enterocolitis Prevention. Mol. Nutr. Food Res. 2019, 63, 1801247. [Google Scholar] [CrossRef]
  46. Burkova, E.E.; Dmitrenok, P.S.; Bulgakov, D.V.; Vlassov, V.V.; Ryabchikova, E.I.; Nevinsky, G.A. Exosomes from human placenta purified by affinity chromatography on sepharose bearing immobilized antibodies against CD81 tetraspanin contain many peptides and small proteins. IUBMB Life 2018, 70, 1144–1155. [Google Scholar] [CrossRef] [Green Version]
  47. Savina, A.; Fader, C.M.; Damiani, M.T.; Colombo, M.I. Rab11 Promotes Docking and Fusion of Multivesicular Bodies in a Calcium-Dependent Manner. Traffic 2005, 6, 131–143. [Google Scholar] [CrossRef]
  48. Hurley, J.H.; Odorizzi, G. Get on the exosome bus with ALIX. Nat. Cell Biol. 2012, 14, 654–655. [Google Scholar] [CrossRef]
  49. Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Pols, M.S.; Klumperman, J. Trafficking and function of the tetraspanin CD63. Exp. Cell Res. 2009, 315, 1584–1592. [Google Scholar] [CrossRef] [PubMed]
  51. Gross, J.C.; Chaudhary, V.; Bartscherer, K.; Boutros, M. Active Wnt proteins are secreted on exosomes. Nat. Cell Biol. 2012, 14, 1036–1045. [Google Scholar] [CrossRef]
  52. Luga, V.; Zhang, L.; Viloria-Petit, A.M.; Ogunjimi, A.A.; Inanlou, M.R.; Chiu, E.; Buchanan, M.; Hosein, A.N.; Basik, M.; Wrana, J.L. Exosomes Mediate Stromal Mobilization of Autocrine Wnt-PCP Signaling in Breast Cancer Cell Migration. Cell 2012, 151, 1542–1556. [Google Scholar] [CrossRef] [Green Version]
  53. Théry, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic analysis of dendritic cell-derived exosomes: A secreted subcellular compartment distinct from apoptotic vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Koh, Y.Q.; Peiris, H.N.; Vaswani, K.; Meier, S.; Burke, C.R.; Macdonald, K.A.; Roche, J.R.; Almughlliq, F.; Arachchige, B.J.; Reed, S.; et al. Characterization of exosomes from body fluids of dairy cows. J. Anim. Sci. 2017, 95, 3893. [Google Scholar] [CrossRef]
  55. Yang, M.; Song, D.; Cao, X.; Wu, R.; Liu, B.; Ye, W.; Wu, J.; Yue, X. 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. [Google Scholar] [CrossRef] [PubMed]
  56. 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]
  57. 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. [Google Scholar] [CrossRef] [PubMed]
  58. Reinhardt, T.A.; Sacco, R.E.; Nonnecke, B.J.; Lippolis, J.D. 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]
  59. Burgoyne, R.D.; Duncan, J.S. Secretion of milk proteins. J. Mammary Gland Biol. Neoplasia 1998, 3, 275–286. [Google Scholar] [CrossRef]
  60. Zeng, B.; Chen, T.; Luo, J.; Xie, M.; Wei, L.; Xi, Q.; Sun, J.; Zhang, Y. Exploration of Long Non-coding RNAs and Circular RNAs in Porcine Milk Exosomes. Front. Genet. 2020, 11, 1–12. [Google Scholar] [CrossRef]
  61. Hata, T.; Murakami, K.; Nakatani, H.; Yamamoto, Y.; Matsuda, T.; Aoki, N. Isolation of bovine milk-derived microvesicles carrying mRNAs and microRNAs. Biochem. Biophys. Res. Commun. 2010, 396, 528–533. [Google Scholar] [CrossRef]
  62. Kosaka, N.; Izumi, H.; Sekine, K.; Ochiya, T. microRNA as a new immune-regulatory agent in breast milk. Silence 2010, 1, 7. [Google Scholar] [CrossRef] [Green Version]
  63. 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, 9, e88843. [Google Scholar] [CrossRef] [PubMed]
  64. 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]
  65. 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] [PubMed] [Green Version]
  66. Chen, T.; Xi, Q.-Y.; Ye, R.-S.; Cheng, X.; Qi, Q.-E.; Wang, S.-B.; Shu, G.; Wang, L.-N.; Zhu, X.-T.; Jiang, Q.-Y.; et al. Exploration of microRNAs in porcine milk exosomes. BMC Genom. 2014, 15. [Google Scholar] [CrossRef] [Green Version]
  67. Mirza, A.H.; Kaur, S.; Nielsen, L.B.; Størling, J.; Yarani, R.; Roursgaard, M.; Mathiesen, E.R.; Damm, P.; Svare, J.; Mortensen, H.B.; et al. Breast Milk-Derived Extracellular Vesicles Enriched in Exosomes From Mothers With Type 1 Diabetes Contain Aberrant Levels of microRNAs. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef]
  68. Chen, Z.; Xie, Y.; Luo, J.; Chen, T.; Xi, Q.; Zhang, Y.; Sun, J. Milk exosome-derived miRNAs from water buffalo are implicated in immune response and metabolism process. BMC Vet. Res. 2020, 16, 123. [Google Scholar] [CrossRef]
  69. Leiferman, A.; Shu, J.; Upadhyaya, B.; Cui, J.; Zempleni, J. Storage of Extracellular Vesicles in Human Milk, and MicroRNA Profiles in Human Milk Exosomes and Infant Formulas. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 235–238. [Google Scholar] [CrossRef]
  70. Kahn, S.; Liao, Y.; Du, X.; Xu, W.; Li, J.; Lönnerdal, B. Exosomal MicroRNAs in Milk from Mothers Delivering Preterm Infants Survive in Vitro Digestion and Are Taken Up by Human Intestinal Cells. Mol. Nutr. Food Res. 2018, 62, 1701050. [Google Scholar] [CrossRef]
  71. 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]
  72. 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, 1–11. [Google Scholar] [CrossRef]
  73. Galley, J.D.; Besner, G.E. The therapeutic potential of breast milk-derived extracellular vesicles. Nutrients 2020, 12, 745. [Google Scholar] [CrossRef] [Green Version]
  74. van Herwijnen, M.J.C.; Driedonks, T.A.P.; Snoek, B.L.; Kroon, A.M.T.; Kleinjan, M.; Jorritsma, R.; Pieterse, C.M.J.; Nolte-‘t Hoen, E.N.M.; Wauben, M.H.M. Abundantly Present miRNAs in Milk-Derived Extracellular Vesicles Are Conserved Between Mammals. Front. Nutr. 2018, 5, 1–6. [Google Scholar] [CrossRef] [PubMed]
  75. Floris, I.; Kraft, J.; Altosaar, I. Roles of MicroRNA across Prenatal and Postnatal Periods. Int. J. Mol. Sci. 2016, 17, 1994. [Google Scholar] [CrossRef] [Green Version]
  76. Meng, F.; Henson, R.; Wehbe-Janek, H.; Smith, H.; Ueno, Y.; Patel, T. The MicroRNA let-7a Modulates Interleukin-6-dependent STAT-3 Survival Signaling in Malignant Human Cholangiocytes. J. Biol. Chem. 2007, 282, 8256–8264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Skotland, T.; Hessvik, N.P.; Sandvig, K.; Llorente, A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J. Lipid Res. 2019, 60, 9–18. [Google Scholar] [CrossRef] [Green Version]
  78. Egea-Jimenez, A.L.; Zimmermann, P. Lipids in Exosome Biology. In Lipid Signaling in Human Diseases; Springer: Cham, Switzerland, 2019; pp. 309–336. [Google Scholar]
  79. Lukic, A.; Ji, J.; Idborg, H.; Samuelsson, B.; Palmberg, L.; Gabrielsson, S.; Rådmark, O. Pulmonary epithelial cancer cells and their exosomes metabolize myeloid cell-derived leukotriene C 4 to leukotriene D 4. J. Lipid Res. 2016, 57, 1659–1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296. [Google Scholar] [CrossRef] [Green Version]
  81. Elsharkasy, O.M.; Nordin, J.Z.; Hagey, D.W.; de Jong, O.G.; Schiffelers, R.M.; Andaloussi, S.L.; Vader, P. Extracellular vesicles as drug delivery systems: Why and how? Adv. Drug Deliv. Rev. 2020. [Google Scholar] [CrossRef]
  82. Hood, J.L. Post isolation modification of exosomes for nanomedicine applications. Nanomedicine 2016, 11, 1745–1756. [Google Scholar] [CrossRef] [Green Version]
  83. Lagassé, H.A.D.; Alexaki, A.; Simhadri, V.L.; Katagiri, N.H.; Jankowski, W.; Sauna, Z.E.; Kimchi-Sarfaty, C. Recent advances in (therapeutic protein) drug development. F1000Research 2017, 6, 113. [Google Scholar] [CrossRef] [Green Version]
  84. Akao, Y.; Iio, A.; Itoh, T.; Noguchi, S.; Itoh, Y.; Ohtsuki, Y.; Naoe, T. Microvesicle-mediated RNA Molecule Delivery System Using Monocytes/Macrophages. Mol. Ther. 2011, 19, 395–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J.A. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef] [PubMed]
  86. Smyth, T.; Kullberg, M.; Malik, N.; Smith-Jones, P.; Graner, M.W.; Anchordoquy, T.J. Biodistribution and delivery efficiency of unmodified tumor-derived exosomes. J. Control. Release 2015, 199, 145–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Smyth, T.J.; Redzic, J.S.; Graner, M.W.; Anchordoquy, T.J. Examination of the specificity of tumor cell derived exosomes with tumor cells in vitro. Biochim. Biophys. Acta Biomembr. 2014, 1838, 2954–2965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Hagiwara, K.; Ochiya, T.; Kosaka, N. A paradigm shift for extracellular vesicles as small RNA carriers: From cellular waste elimination to therapeutic applications. Drug Deliv. Transl. Res. 2014, 4, 31–37. [Google Scholar] [CrossRef] [Green Version]
  89. Aqil, F.; Munagala, R.; Jeyabalan, J.; Gupta, R.C. Abstract 5407: Milk derived exosomes: Scalable source of biologically active drug delivery nanoparticles. In Proceedings of the Cancer Chemistry; American Association for Cancer Research: Philadelphia, PA, USA, 2014; p. 5407. [Google Scholar]
  90. Somiya, M.; Yoshioka, Y.; Ochiya, T. Biocompatibility of highly purified bovine milk-derived extracellular vesicles. J. Extracell. Vesicles 2018, 7. [Google Scholar] [CrossRef] [Green Version]
  91. Badawy, A.A.; El-Magd, M.A.; AlSadrah, S.A. Therapeutic Effect of Camel Milk and Its Exosomes on MCF7 Cells In Vitro and In Vivo. Integr. Cancer Ther. 2018, 17. [Google Scholar] [CrossRef] [Green Version]
  92. Lin, D.; Chen, T.; Xie, M.; Li, M.; Zeng, B.; Sun, R.; Zhu, Y.; Ye, D.; Wu, J.; Sun, J.; et al. Oral Administration of Bovine and Porcine Milk Exosome Alter miRNAs Profiles in Piglet Serum. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef]
  93. Aqil, F.; Munagala, R.; Jeyabalan, J.; Agrawal, A.K.; Gupta, R. Exosomes for the Enhanced Tissue Bioavailability and Efficacy of Curcumin. AAPS J. 2017, 19, 1691–1702. [Google Scholar] [CrossRef]
  94. 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]
  95. Carobolante, G.; Mantaj, J.; Ferrari, E.; Vllasaliu, D. Cow Milk and Intestinal Epithelial Cell-Derived Extracellular Vesicles as Systems for Enhancing Oral Drug Delivery. Pharmaceutics 2020, 12, 226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Allison, A.C.; Cacabelos, R.; Lombardi, V.R.M.; Álvarez, X.A.; Vigo, C. Celastrol, a potent antioxidant and anti-inflammatory drug, as a possible treatment for Alzheimer’s disease. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2001, 25, 1341–1357. [Google Scholar] [CrossRef]
  97. Chang, F.-R.; Hayashi, K.; Chen, I.-H.; Liaw, C.-C.; Bastow, K.F.; Nakanishi, Y.; Nozaki, H.; Cragg, G.M.; Wu, Y.-C.; Lee, K.-H. Antitumor Agents. 228. Five New Agarofurans, Reissantins A−E, and Cytotoxic Principles from Reissantia b uchananii. J. Nat. Prod. 2003, 66, 1416–1420. [Google Scholar] [CrossRef] [PubMed]
  98. Aqil, F.; Kausar, H.; Agrawal, A.K.; Jeyabalan, J.; Kyakulaga, A.-H.; Munagala, R.; Gupta, R. Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp. Mol. Pathol. 2016, 101, 12–21. [Google Scholar] [CrossRef] [PubMed]
  99. Munagala, R.; Aqil, F.; Jeyabalan, J.; Agrawal, A.K.; 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]
  100. Aqil, F.; Jeyabalan, J.; Agrawal, A.K.; 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]
  101. Betker, J.L.; Angle, B.M.; Graner, M.W.; Anchordoquy, T.J. The Potential of Exosomes From Cow Milk for Oral Delivery. J. Pharm. Sci. 2019. [Google Scholar] [CrossRef]
  102. Li, D.; Yao, S.; Zhou, Z.; Shi, J.; Huang, Z.; Wu, Z. Hyaluronan decoration of milk exosomes directs tumor-specific delivery of doxorubicin. Carbohydr. Res. 2020, 493, 108032. [Google Scholar] [CrossRef]
  103. Saneja, A.; Arora, D.; Kumar, R.; Dubey, R.D.; Panda, A.K.; Gupta, P.N. CD44 targeted PLGA nanomedicines for cancer chemotherapy. Eur. J. Pharm. Sci. 2018, 121, 47–58. [Google Scholar] [CrossRef]
  104. Komine-Aizawa, S.; Ito, S.; Aizawa, S.; Namiki, T.; Hayakawa, S. Cow milk exosomes activate NK cells and γδT cells in human PBMCs in vitro. Immunol. Med. 2020, 1–10. [Google Scholar] [CrossRef]
  105. Shandilya, S.; Rani, P.; Onteru, S.K.; Singh, D. Small Interfering RNA in Milk Exosomes Is Resistant to Digestion and Crosses the Intestinal Barrier In Vitro. J. Agric. Food Chem. 2017, 65, 9506–9513. [Google Scholar] [CrossRef] [PubMed]
  106. 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. [Google Scholar] [CrossRef] [PubMed]
  107. Downer, M.K.; Batista, J.L.; Mucci, L.A.; Stampfer, M.J.; Epstein, M.M.; Håkansson, N.; Wolk, A.; Johansson, J.-E.; Andrén, O.; Fall, K.; et al. Dairy intake in relation to prostate cancer survival. Int. J. Cancer 2017, 140, 2060–2069. [Google Scholar] [CrossRef] [PubMed]
  108. Tate, P.L.; Bibb, R.; Larcom, L.L. Milk Stimulates Growth of Prostate Cancer Cells in Culture. Nutr. Cancer 2011, 63, 1361–1366. [Google Scholar] [CrossRef]
  109. 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. Biol. Sci. 2012, 8, 118–123. [Google Scholar] [CrossRef]
  110. Baddela, V.S.; Nayan, V.; Rani, P.; Onteru, S.K.; Singh, D. Physicochemical Biomolecular Insights into Buffalo Milk-Derived Nanovesicles. Appl. Biochem. Biotechnol. 2016, 178, 544–557. [Google Scholar] [CrossRef]
  111. 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. 2017, 147, 3–10. [Google Scholar] [CrossRef] [Green Version]
  112. Mishra, S.; Lin, C.-L.; Huang, T.H.-M.; Bouamar, H.; Sun, L.-Z. MicroRNA-21 inhibits p57Kip2 expression in prostate cancer. Mol. Cancer 2014, 13, 212. [Google Scholar] [CrossRef] [Green Version]
  113. Li, S.; Yang, X.; Yang, J.; Zhen, J.; Zhang, D. Serum microRNA-21 as a potential diagnostic biomarker for breast cancer: A systematic review and meta-analysis. Clin. Exp. Med. 2016, 16, 29–35. [Google Scholar] [CrossRef]
  114. Wang, G.; Wang, L.; Sun, S.; Wu, J.; Wang, Q. Quantitative Measurement of Serum MicroRNA-21 Expression in Relation to Breast Cancer Metastasis in Chinese Females. Ann. Lab. Med. 2015, 35, 226. [Google Scholar] [CrossRef]
  115. Worster, D.T.; Schmelzle, T.; Solimini, N.L.; Lightcap, E.S.; Millard, B.; Mills, G.B.; Brugge, J.S.; Albeck, J.G. Akt and ERK Control the Proliferative Response of Mammary Epithelial Cells to the Growth Factors IGF-1 and EGF Through the Cell Cycle Inhibitor p57Kip2. Sci. Signal. 2012, 5, ra19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Reif, S.; ElbaumShiff, Y.; 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. [Google Scholar] [CrossRef] [PubMed]
  117. Dana, H.; Chalbatani, G.M.; Mahmoodzadeh, H.; Karimloo, R.; Rezaiean, O.; Moradzadeh, A.; Mehmandoost, N.; Moazzen, F.; Mazraeh, A.; Marmari, V.; et al. Molecular Mechanisms and Biological Functions of siRNA. Int. J. Biomed. Sci. 2017, 13, 48–57. [Google Scholar] [PubMed]
  118. Behlke, M.A. Chemical Modification of siRNAs for In Vivo Use. Oligonucleotides 2008, 18, 305–320. [Google Scholar] [CrossRef] [Green Version]
  119. Jackson, A.L. Position-specific chemical modification of siRNAs reduces ‘off-target’ transcript silencing. RNA 2006, 12, 1197–1205. [Google Scholar] [CrossRef] [Green Version]
  120. Amarzguioui, M. Tolerance for mutations and chemical modifications in a siRNA. Nucleic Acids Res. 2003, 31, 589–595. [Google Scholar] [CrossRef] [Green Version]
  121. Tatiparti, K.; Sau, S.; Kashaw, S.; Iyer, A. siRNA Delivery Strategies: A Comprehensive Review of Recent Developments. Nanomaterials 2017, 7, 77. [Google Scholar] [CrossRef] [Green Version]
  122. Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and challenges of extracellular vesicle-based drug delivery system: Considering cell source. Drug Deliv. 2020, 27, 585–598. [Google Scholar] [CrossRef] [Green Version]
Table 1. Highly represented proteins of milk exosomes.
Table 1. Highly represented proteins of milk exosomes.
Highly Presented ProteinsNumber of Proteins Described in a PaperSource of Milk Exosomes, RefMethod of Analysis
Butyrophilin, κ-casein, lactadherin, xanthine dehydrogenase94Bovine [54]LC-MS/MS of tryptic hydrolysates
Angiogenin-1, lactoferrin, lactoperoxidase sulfhydryl oxidase 920Bovine [55]LC-MS/MS of tryptic hydrolysates with iTRAQ
Butyrophilin, CD36, complement component 3, fatty acid synthase, lactadherin, lactotransferrin, low-density lipoprotein receptor-related protein 2, polymeric immunoglobulin receptor, xanthine dehydrogenase 1372 Bovine [56]LC-MS/MS of tryptic hydrolysates
α-casein, butyrophilin, fatty acid-binding protein, lactadherin, α-lactalbumin, β-lactoglobulin, xanthine dehydrogenase 1879Bovine [57]LC-MS/MS of tryptic hydrolysates
Adipophilin, butyrophilin, lactadherin, xanthine oxidase 2107Bovine [43] LC-MS/MS of tryptic hydrolysates
Butyrophilin, lactadherin, fatty acid synthase, xanthine dehydrogenase 2299Bovine [58] LC-MS/MS of tryptic hydrolysates with iTRAQ
Actin, butyrophilin, lactadherin, lactoferrin, β-lactoglobulin8Horse [11]MALDI-TOF-MS/MS of tryptic hydrolysates after 2D-Electrophoresis
CD36, α-enolase, fatty acid synthase, lactadherin, lactotransferrin, polymeric-Ig-receptor, Rab GDP dissociation inhibitor, syntenin-1, xanthine dehydrogenase 73Human [5]LC-MS/MS of tryptic hydrolysates
β-Casein, lactoferrin, serum albumin polymeric Ig receptor, tenascin, xanthine dehydrogenase 115Human [41]LC-MS/MS of tryptic hydrolysates
Annexins, CD9 CD63, CD81, flotillin, G-protein subunits, lactadherin, Rab, Ras-related proteins, syntenin2698Human [44]LC-MS/MS of tryptic hydrolysates
Albumin, ceruloplasmin, complement C, α-glucosidase, fibronectin, lactotransferrin, thrombospondin 571Porcine [42]LC-MS/MS of tryptic hydrolysates after SDS PAGE
Table 2. Highly represented microRNA of milk exosomes.
Table 2. Highly represented microRNA of milk exosomes.
Highly Presented microRNANumber of microRNAs Described in a PaperSource of Milk Exosomes, RefMethod of Analysis
2478, 1777b, 1777a, let-7b, 1224, 2412, 2305, let-7a, 200c, 14179Bovine [65]Microarray
148a, let-7c, let-7a-5p, 26a, let-7f, 30a-5p, 30d372Buffalo [68]RNA seq
30d-5p, let-7b-5p, let-7a-5p, 125a-5p, 21-5p, 423-5p, let-7g-5p, let-7f-5p, 30a-5p, 146b-5p219Human [69]RNA seq
22-3p, 148a-3p, 141-3p, 181a-5p, 320a, 378a-3p, 30d-5p, 30a-5p, 26a-5p, 191-5p 308Human 1 [66] RNA seq
let-7a-5p, 148a-3p, 146b-5p, let-7f-5p, let-7g-5p, 21-5p, 26a-5p, 30d-5p631Human [70]RNA seq
148a-3p, 30b-5p, let-7f-5p, 146b-5p, 29a-3p, let-7a-5p, 141-3p, 182-5p, 200a-3p, 378-3p 602Human [67]RNA seq
let-7b-5p, 92a-3p, 148a-3p, 30a-5p, let-7a-5p, 181a-5p, let-7i-5p, let-7f-1/2-5p, let-7g-5, 200a-3p1191Panda [12]RNA seq
148a-3p, 182-5p, 200c-3p, 25-3p, 30a-5p, 30d-5p, 574-3p234Porcine [7]RNA seq
148a, let-7b, let-7a, 21, let-7c, let-7i, 26a, let-7f, 125b, 14384Sheep [14]RNA seq
1 Preterm delivery.
Table 3. Genes regulated by a highly abundant microRNA of milk exosomes.
Table 3. Genes regulated by a highly abundant microRNA of milk exosomes.
microRNA(s)Gene(s) Targeting with microRNABiological Function(s) of the microRNA Targets, Ref
22-3pTranscription factor 7 of Wnt pathwayRegulation of gluconeogenesis, insulin resistance [72]
25-3pKLF4 (Krüppel-like factor 4)Development of the immune system [7]
30a-5pP53, DRP1 (dynamin-related protein 1), GALNT7 (GalNAc transferase 7) Mitochondrial fission, cellular invasion, immunosuppression, synthesis of interleukin (IL)-10 [7]
30d-5pGalNAc transferasesInflammatory processes [73]
148-3pNF-κB (transcription factor)Decrease of the immune response [74]
148a 1 DNMT1 Epigenetic regulation [71]
let-7 familyInsulin-PI3K-mTOR signaling pathwayGlucose tolerance and insulin sensitivity [75]
let-7a-5p
let-7b-5p
TRIM71, IL6-induced signal activation of transcription, Stem cells proliferation, fetal development [74], activation of metalloproteinases [76]
1 The most expressed miRNA of human milk.
Table 4. Biologically and therapeutically significant molecules delivered with milk exosomes.
Table 4. Biologically and therapeutically significant molecules delivered with milk exosomes.
Molecule DeliveredSolubility in WaterSource of Milk Exosomes, RefCell Lines Used for Delivery
AnthocyaninsSolubleBovine [99,100]A549 1, H1299 1, MDA-MB-231 2, MCF7 2, PANC1 3, Mia PaCa2 3, PC3 4, DU145 4, HCT116 5, OVCA432 6, OVCA433 6, A2780 6, A2780/CP70 6
CelastrolInsolubleBovine [98]A549 1, H1299 1
CurcuminInsolubleBovine [94,95,98]Caco-2 5, H1299 1, A549 2, HeLa 7, MDA-MB-231 2, T47D 2
DocetaxelInsolubleBovine [18]A549 1, H1299 1, MB-231 2, T47D 2, Beas-2B 8
DoxorubicinSoluble Bovine [102]A549 1, MDA-MB-231 2, MCF-7 2, HEK293 9
PaclitaxelInsolubleBovine [17,18]A549 1, H1299 1, MB-231 2, T47D 2, Beas-2B 8
siRNASolubleBovine [20,105]A549 1, H1299 1, MDA-MB-231 2, MCF7 2, PANC1 3, Mia PaCa2 3, Caco-2 5, A2780 6
Withaferin AInsolubleBovine [18]A549 1, H1299 1, MB-231 2, T47D 2, Beas-2B 8
1 lung cancer, 2 breast cancer, 3 pancreatic cancer, 4 prostate cancer, 5 colon cancer, 6 ovarian cancer, 7 cervical cancer, 8 normal bronchial epithelium, 9 normal kidney embryonic cells.

Share and Cite

MDPI and ACS Style

Sedykh, S.; Kuleshova, A.; Nevinsky, G. Milk Exosomes: Perspective Agents for Anticancer Drug Delivery. Int. J. Mol. Sci. 2020, 21, 6646. https://doi.org/10.3390/ijms21186646

AMA Style

Sedykh S, Kuleshova A, Nevinsky G. Milk Exosomes: Perspective Agents for Anticancer Drug Delivery. International Journal of Molecular Sciences. 2020; 21(18):6646. https://doi.org/10.3390/ijms21186646

Chicago/Turabian Style

Sedykh, Sergey, Anna Kuleshova, and Georgy Nevinsky. 2020. "Milk Exosomes: Perspective Agents for Anticancer Drug Delivery" International Journal of Molecular Sciences 21, no. 18: 6646. https://doi.org/10.3390/ijms21186646

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop