Next Article in Journal
Interaction between Polyphenolic Antioxidants and Saccharomyces cerevisiae Cells Defective in Heavy Metal Transport across the Plasma Membrane
Next Article in Special Issue
A Comparative View on the Oviductal Environment during the Periconception Period
Previous Article in Journal
Characterization of Bacterial Cellulose-Based Wound Dressing in Different Order Impregnation of Chitosan and Collagen
Previous Article in Special Issue
Determining the Molecular Background of Endometrial Receptivity in Adenomyosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Biological Function of Extracellular Vesicles during Fertilization, Early Embryo—Maternal Crosstalk and Their Involvement in Reproduction: Review and Overview

by
Emanuele Capra
1 and
Anna Lange-Consiglio
2,3,*
1
Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche IBBA CNR, 26900 Lodi, Italy
2
Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, 26900 Lodi, Italy
3
Centro Clinico-Veterinario e Zootecnico-Sperimentale di Ateneo, Università degli Studi di Milano, 26900 Lodi, Italy
*
Author to whom correspondence should be addressed.
Biomolecules 2020, 10(11), 1510; https://doi.org/10.3390/biom10111510
Submission received: 28 September 2020 / Revised: 29 October 2020 / Accepted: 31 October 2020 / Published: 4 November 2020
(This article belongs to the Special Issue Biomolecules in Maternal–Embryo Communication in Implantation)

Abstract

:
Secretory extracellular vesicles (EVs) are membrane-enclosed microparticles that mediate cell to cell communication in proximity to, or distant from, the cell of origin. Cells release a heterogeneous spectrum of EVs depending on their physiologic and metabolic state. Extracellular vesicles are generally classified as either exosomes or microvesicles depending on their size and biogenesis. Extracellular vesicles mediate temporal and spatial interaction during many events in sexual reproduction and supporting embryo-maternal dialogue. Although many omic technologies provide detailed understanding of the molecular cargo of EVs, the difficulty in obtaining populations of homogeneous EVs makes difficult to interpret the molecular profile of the molecules derived from a miscellaneous EV population. Notwithstanding, molecular characterization of EVs isolated in physiological and pathological conditions may increase our understanding of reproductive and obstetric diseases and assist the search for potential non-invasive biomarkers. Moreover, a more precise vision of the cocktail of biomolecules inside the EVs mediating communication between the embryo and mother could provide new insights to optimize the therapeutic action and safety of EV use.

1. Introduction

In mammals, the female reproductive trait, comprising ovaries, oviducts and uterus, plays a crucial role in the regulation of early and late reproductive events and provides the optimal environment for embryonic development. Secretory extracellular vesicles (EVs) are important for cell to cell communication, as they have a paracrine function being able to diffuse over a relatively short distance and produce a local action. It has been largely demonstrated that EV-associated activity is fundamental to reproductive success, mediating the fine-tuning of cellular cross-talk in the reproductive system, promoting embryo implantation and assisting successful pregnancy [1]. Especially in reproduction fields, many early studies used low-specific EV isolation methods that often co-isolated other soluble molecules such as growth factors, cytokines and metabolites, making difficult to attribute the activity specifically to EVs and not instead to low amount of other highly active soluble mediators. Extracellular vesicles (EVs) are membrane bound organelles which can convey information between cells through the transfer of functional protein and genetic information to alter phenotype and function of recipient cells [2,3]. Many recent reviews highlight the role of EVs in oogenesis, oocyte maturation and fertilization, embryo-maternal cross talk in the oviduct and embryo implantation [1,4,5,6,7,8,9,10,11,12,13]. In this review, the recent knowledge on maternal-fetal communication, with special reference to the role of EVs during early interaction between embryonic and maternal tissues are reviewed. At first, we summarize the methods used for EV isolation and their molecular and functional characterization to underline the complexity of multi-signaling response in EV-mediated cell to cell communication. Then, we examine the key role of EVs in early reproductive events and embryo-maternal cross talk, focusing on the molecular cargo of the EVs that transmits messages to different compartments of the reproductive tract and are involved in embryo-maternal cross-talk. Finally, we analyze EVs and their cargo for potential novel diagnostic and therapeutic applications in animal and human reproductive medicine (Figure 1).

2. Extracellular Vesicles and Biogenesis

Membrane-enclosed microparticles were first isolated from biological materials such as animal platelet-free plasma [14], human seminal plasma [15] and from a variety of mammalian cell lines [16]. The isolated vesicles were assumed to originate from outward budding of plasma membranes. It was not until years later that a novel pathway involving active vesicle secretion was described in reticulocytes and the term exosomes was used to define intracellular endosome vesicles released by exocytosis [17]. Although EVs have been isolated from all types of organisms including Archaea [18], Bacteria [19] and Eukarya [20], in animals they were classified into two main categories: exosomes and microvesicles depending on their size and biogenesis. The smallest exosomes (ranging in size between 50 and 150 nm in diameter) originate from the multivesicular endolytic compartment by the fusion of multivesicular endosome with the plasma membrane [21,22]. Microvesicles (ranging in size between 50 nm and 1,000 nm) are shed by an active outward budding mechanism mediated by reorganization of the actin-myosin cytoskeleton [23,24].

3. Methods for EV Isolation and Characterization

Cells produce and release a heterogeneous spectrum of EVs differing in size and chemical and physical characteristics. Many methods based on centrifugation, filtration, precipitation and affinity interaction have been used to isolate EVs [25]. The isolation of EVs from complex body fluids such as blood or follicular fluid often requires a combination of different techniques such as differential centrifugation, density gradient centrifugation, filtration, affinity-based method precipitation with polymers such as polyethylene glycol and size exclusion chromatography [25,26,27,28,29,30,31]. Recently, a new type of small (<50 nm), non-membranous nanoparticle named exomeres was separated from various secreted vesicles using asymmetric flow field-flow fractionation technology and ultracentrifugation [32,33]. Isolation efficiency is dependent on the type of sample and the research purposes and procedures must be carefully standardized [34].
Each method present advantages and disadvantages and should be carefully chosen based on sample type, downstream application and scientific question. Some indications are reported in the guidelines presented by international EV body, ISEV [34,35] and summarized in Table 1.
All procedures include methods for evaluation of vesicle morphology, size distribution and concentration so that the purified EVs can be correctly identified. A topographical EV view is obtained by direct imaging methods including scanning (SEM),transmission electronic microscopy (TEM) and cryo-electron microscopy (cryo-EM) that provide high resolution imaging and a more precise evaluation of vesicular size [36,37]. However, both techniques require laborious sample preparation procedures that limit sample throughput. Methods based on measurement of the Brownian motion of suspended particles include nanoparticle tracking analysis (NTA) and dynamic light scattering (DLS), or flow cytometry (FC) and tunable resistive pulse sensing (TRPS) methods can be used to measure EV concentration in solution or for determination of the EV size ranges when high-throughput information is desired. However, the detection of EVs becomes difficult when large vesicles are present in dynamic light scattering methods [38,39]. Noble et al. [40] reviewed a comparison between electron microscopy and optical methods for EV detection. Isolated vesicles are likely to be a combination of microvesicles and exosomes varying in size [40]. Thus, further biochemical characterization is needed to provide information on EV composition. Extracellular vesicle preparation identity and purity must be evaluated by detecting the presence of anchored protein localized at the external membrane such as transmembrane (CD63, CD81, CD82, CD47) or GPI-anchored protein (CD73) and the absence of major constituent proteins of non EV structures that co-isolate with EVs such as albumin (ALB) and apolipoproteins A1/2 and B (APO1/2, APOB). In addition, to evaluate EV integrity of lipid bilayers the presence of cytosolic protein (ALIX, heat shock proteins HSPs) should be taken into consideration [35]. Subtype characterization can be obtained by detecting the presence of proteins associated to different intracellular compartments. Extracellular vesicles subtypes separation from peripheral blood confirmed the specific isolation of microvescicles using specific antibody targeting proteins, reflecting their biogenesis such as tubulin, actinin-4 or mitofilin for microvesicles and tetraspanin antibodies (e.g., CD9, CD81) for exosomes [41].Protein markers for EV characterization are detected using different technologies. Western blotting and enzyme-linked immunosorbent assay (ELISA) are the most commonly used method to evaluate the presence of EV associated markers. In general, both techniques require a large sample volume and long processing. The limit of detection is similar for both methods, but ELISA can be easily scaled up for higher-throughput measurements. In alternative, EV protein composition can be assessed by mass spectrometry, that provides quantitative and comparative EV proteomic characterization, although requires significant preparatory and processing time. Surface protein marker can be detected using other methods such as small particle flow cytometry, surface plasma resonance. Methods for EV protein characterization were extensively reviewed by Shao et al. [42].
Extracellular vesicles represent an important mode of intercellular communication by facilitating the horizontal cell to cell transfer of lipids, proteins, RNA species and DNA fragments. However, the functionality and characterization of EV molecular cargo is not always easy to interpret due to the presence of different isolate subtypes and the inability to further separate and to stratify these into distinct sub-populations [43,44].

4. Extracellular Vesicle Composition and their Molecular Cargo Function

Lipidomics and proteomics have identified about 2000 lipid and 3500 protein species isolated from EVs secreted by different cell sources [45,46]. Extracellular vesicle lipid composition is specific for cellular type. Extracellular vesicles can transfer lipid mediators that regulate several pathologic processes including inflammation, tumor development, and atherogenesis [47]. Extracellular vesicles contain a subset of cellular proteins derived from plasma membranes, endosomes, and cytosol that are consistent with their biogenesis and cellular type. Among them, specific proteins can be used to distinguish between different EV classes and subtypes [48]. Extracellular vesicle surface proteins, such as glycan binding proteins, are probably used by the vesicle for cell specific targeting. The presence of amyloid-β (A4) protein outside EVs mediates the destination of EVs from neuroblastoma cell to neurons in the central nervous system. At the same time, the expression of some proteins such as beta amyloid peptide, synuclein and TAR DNA-binding protein 43 (TDP43) changes in neuronal disease and can be used as a marker for progression [49]. Extracellular vesicle proteins can also mediate cellular communication. Surface proteins present on EVs released by the inner cell mass such as laminin and fibronectin can interact with integrins on the surfaces of the trophoblast promoting embryo implantation [50].
The emerging role of cell to cell communication is mediated by EV-associated small non-coding regulatory RNAs called microRNAs (miRNAs) and other non-coding RNAs by acting as “messengers” shuttled between different cell types. In fact, miRNAs can be specifically compartmentalized inside vesicles and released outside cells, as previously reported in mesenchymal stromal cells in human and equine studies [51,52]. However, nonselective miRNA loading and secretion has also been reported [53,54]. The relative contributions of passive and active packaging of RNAs into EVs is not fully understood [55]. In addition, stoichiometric analysis of EV miRNA cargo indicate that EVs isolated from different tissues do not contain many copies of miRNAs [56]. This unexpected result may be explained by the fact that isolated EVs represent a heterogeneous population of vesicles with different physical and molecular properties [57,58].
Many aspects of how miRNAs are selectively encapsulated in EVs and can mediate cell to cell communication are still not understood [59]. Molecular profiling of EVs surface revealed specific molecular composition that mediate interactions at the surface of extracellular vesicles [60,61]. It is likely that EVs recognize and transfer their molecular content such as miRNAs proteins and lipids, through an active internalization process mediated by various endocytic mechanisms such as clathrin dependent and caveolae dependent endocytosis and lipid raft mediated uptake. In fact, antibodies against specific proteins such as tetraspanins, integrins, immunoglobulins, proteoglycans, lectins block vesicular entry into recipient cells [62,63].
Other nucleic acids, including mRNAs and DNAs, are important in regulation of gene expression but their role in the transfer of cell to cell information remains controversial. Transcripts have been observed to be delivered by EVs but rapidly degraded, whereas plasmid DNA can only be delivered by EVs for correct functioning in recipient cells [64]. Recently, EVs were observed to mediate horizontal gene transfer during genome editing. The use of the CRISPR-Cas9 system in NIH-3T3 mouse cells showed an unexpected insertion of bovine DNA, derived from satellite DNA elements, that was mediated by EVs present in the cell culture medium [65]. Double-stranded DNA in EVs has also been used in medical diagnostics as a biomarker for cancer [66,67].

5. Extracellular Vesicle in Animal Reproduction and Embryo-Maternal Cross-Talk

Cell communication is fundamental for several molecular processes that allow populations of cells to exchange information with one another to aid specialization in tissue or coordinate multiple cross-organ interaction.
Extracellular vesicles have been identified as one of the key players in regulating temporal sequences and spatial interaction, cell-to cell signaling in all events in sexual reproduction (i.e., gametogenesis, fertilization and embryogenesis) and in embryo-maternal cross-talk (Figure 2 and Table 2).
Extracellular vesicles in seminal fluids were observed to modulate sperm capacitation in man [82] and pigs [83] and to influence female physiology by modulating immune-related gene expression in the porcine endometrium [68]. In return, oviductal EV (oEV) secretion was observed to regulate sperm motility and capacitation in mouse [69,70], cat [71] and bovine [72] sperm and EVs from avian uterine fluid may play an essential role in the preservation of sperm functions [81]. During oogenesis, follicle growth and oocyte maturation required a constant signal exchange in which there is a mutual recognition of diffusible or EV-mediated molecules produced by somatic cells (granulosa, cumulus and theca) and by germ cells (oocytes) present in the follicular fluids (FF). Extracellular vesicles isolated from FF were first observed and characterized in horse [84], human [85] and bovine [86] studies. Follicular fluid comprises a heterogeneous EV population secreted by granulosa, cumulus and somatic follicular cells with functions related to control of steroidogenesis [85,87]. Extracellular vesicles from FF were able to modulate bovine embryo development in vitro [75].
The successful of pregnancy requires a molecular dialogue between the embryo and the female reproductive tract that starts at the oviduct and continues until the formation of placenta. The first known molecules in this paracrine communication were the cytokines and growth factors, such as interleukin-1ß (IL-1ß), heparin-binding epidermal growth factor (HB-EGF), integrins and leukemia inhibitory factor (LIF) that act synergistically in the embryo-maternal crosstalk. The IL-1 increases the expression of molecules of adhesion of epithelial cells regulating the endometrial receptivity [88] and stimulating angiogenesis to promote the embryonic growth [89]. The HB-EGF is one of the members of epidermal growth factor family and, through the HB-EGF receptors on the surface of embryo and endometrium, facilitates the process of implantation and promotes the development of blastocyst [90,91]. The expression levels of IL-1β and HB-EGF are higher in implantation phase and are positively correlated with the levels of estradiol and progesterone [92], showing their important role in this phase. Integrins are adhesion molecules and constitute a system of cell-cell and cell-matrix interaction. During implantation, the integrins α6β1 and α7ß1 of blastocyst bind to laminin of the basement membrane of endometrium, while the integrin α4β1 of endometrium interferes with trophoblastic fibronectin. In this way, this new set of integrins promotes the interaction between the trophectoderm and the endometrium and then the implantation [93]. The LIF has different role in embryo implantation and it is produced by both the blastocyst and the endometrium: increases the expression of EGF and implantation genes in receptive endometrium; regulates growth and development of the embryo; increases the production of cytokines and prostaglandins; stimulates trophoblast cell differentiation and increases trophoblast capability to invade the uterine stroma and it is involved in recruitment of specific cohort of leucocytes which participates in uterine inflammatory response during implantation. The action of LIF is due, for example, to the regulation of expression of adhesion molecules such as L-selectins, E-cadherins and tight junction proteins or to the activation of signal transducer and activator of transcription 3 (STAT3) phosphorylation, or to production of prostaglandin E (PGE2) stimulating COX-2 and microsomal PGE synthase-1 (mPGES-1) enzymes expression that are involved in PGE synthesis. Due to its important role, this factor is proposed as non-hormonal contraceptive [94]. The LIF, with other cytokines, chemokines, and chemokine receptors, is present in EVs too [95] but while the soluble secretome is involved on mouse embryo growth, the EVs are implicated on implantation and embryo development and may be implicated in the pathophysiology of implantation failure in infertility [7].
Indeed, the EVs mediate two-way trafficking of molecules for embryo-maternal communication. During embryo migration from the oviduct to the uterus, the embryo undergoes distinct metabolic stages (cleavage, morula, blastula, gastrula). Labelled in vivo EV preparations from bovine oviduct flushing were internalized by in vitro produced embryos [73]. Both in vivo oEVs and in vitro EVs isolated from bovine oviduct epithelial cell culture were shown to exert a positive effect on development and quality of in vitro produced cattle embryos [73,74]. Although a reciprocal regulation mediated by EVs from the embryo has not yet been documented, vesicles from the embryo can regulate embryo development and implantation. In mice, laminin and fibronectin present in EVs derived from the preimplantation blastocyst inner cell mass (ICM), that gives rise to a whole embryo, interact with integrins on the cell surface of trophectoderm (TE) cells, increasing efficiency of blastocyst implantation [50]. Extracellular vesicles exchange in the uterine environment was reported in vivo whereas fluorescent labelled EVs isolated from sheep uterine fluid were internalized in the conceptus trophectoderm and uterine epithelia [96]. During implantation, a synergistic endometrial-blastocyst crosstalk in the maternal uterine environment is necessary for maternal recognition and successful pregnancy [1]. Intrauterine bovine EVs are required for conceptus implantation and can regulate blastocyst development [79,80]. In human, endometrial-derived EVs were observed to be internalized in HTR8 trophoblast cells and in spheroid model of trophectoderm and enhance cell adhesion capabilities [10,11]. In response, trophoblast cells form the interface with the maternal cells and the fetus and can secrete EVs to enable the fetus to interact with uterine endothelial cells. Conceptus-derived EVs containing Interferon tau (IFN-t) isolated from pregnant ewes were able to modulate expression of genes in bovine endometrial epithelial cells (EECs) [77]. Extracellular vesicles derived from porcine trophectoderm-cells induce aortic endothelial cell proliferation providing evidence of their ability to stimulate angiogenesis [76]. Finally, the placenta secretes EVs carrying proteins involved in immune-modulation which is essential for success of pregnancy [97,98]. In vitro, human placental cytotrophoblast cells are observed to respond to hypoxia by modifying the bioactivity of EVs, thereby, promoting the migration of extravillous trophoblasts into the decidua and myometrium and establishing placental perfusion [78]. Placenta-derived EVs are known to contain different pro-inflammatory molecules that potentially can modulate the local endometrial immune system and mediate local immunotolerance of the fetus to evade the maternal immunosurveillance [99]. In fact, cytotrophoblasts from villous release EVs that are observed to reduce the inflammatory response in peripheral blood mononuclear cells (PBMCs) [100]. Interestingly, a concurrent variation of pro-inflammatory state and dysregulation of placenta-derived EVs and total EVs is observed in some complications of pregnancy such as preeclampsia [101]. To note, EVs isolated from preeclampsia patients also inhibit angiogenesis of human endothelial cells [102], suggesting an active role in cell to cell communication for the maintenance of the correct physiological state during pregnancies.

6. Extracellular Vesicle Molecular Cargo in Animal Reproduction and Embryo-Maternal Cross-Talk

The biological function of EVs in regulating early embryo-maternal interaction results from the transfer of critical molecular cargos to distant or neighboring recipient cells or tissues. Although the latest high-throughput analytical platforms can perform detailed analysis of the lipid, glycolic, proteic and nucleic acid content in EVs [103], it is still difficult to establish the molecular basis of EV cargo regulating embryo-maternal communication because of the many variables associated with EV isolation and characterization. Mass spectrometry analysis shows significant differences in the protein content of in vitro and in vivo EVs secreted from the oviduct [73], and between the protein cargo of placental EVs isolated using different procedures [104]. Isolation of different-sized fractions of EVs from human first-trimester placentae, by differential ultracentrifugation, showed that although many proteins involved in vesicle transport and inflammation are shared between different fractions, some proteins related to vesicle internalization, membrane cofactor and minor histocompatibility antigens, are specific for different-sized EVs. Therefore, it is likely that different sized EVs can interact with different maternal cells mediating multiple messages during feto-maternal communication in early pregnancy [105]. In addition, the overall EV molecular cargo in bovine oviductal fluid changes in function in different stages of the estrous cycle, suggesting that EV composition is under hormonal control [106]. A recent review by Almiñana and Bauersachs, which compared the protein and miRNA content in oEVs, based on literature available data, found only a partial overlap of EV molecular cargo between different species probably because of different EV isolation and characterization methods [5].
Several proteins in EVs regulate different reproductive functions promoting fertilization and embryo implantation and growth. Extracellular vesicles released into the murine uterine luminal fluid deliver sperm adhesion molecule 1 (SPAM1) to sperm membranes, enhancing sperm fertility [107]. Extracellular vesicles from the oviduct transfer plasma membrane Ca2+ ATPase 4 (PMCA4) protein to sperm to enhance Ca2+ efflux in mice [83]. Embryo EVs transport progesterone-induced-blocking factor 1 PIBF that increases IL-10 production in maternal lymphocytes, sustaining immune responses during pregnancy [108]. In return, placental tissue from the first trimester releases EVs containing proteins with roles in regulating and modulating T-cell activity to modify the maternal immunological environment, such as Fas ligand, TNF-related apoptosis-inducing ligand TRAIL, programmed death-ligand 1PD-L1, B7 homolog 1 B7-H1, B7 homolog 3 B7-H3 and human leukocyte antigen G 5 HLA-G5 [109].
MicroRNAs encapsulated in secreted EVs play a significant role in intercellular communication reaching target cells and regulating mRNA and protein expression. Extracellular vesicles transferred from the embryo to the mother, or vice versa, transport miRNAs that target specific pathways for fertilization, embryo implantation and fetal development. Following hormone stimulation, in silico bioinformatic analysis showed that miRNA content, which potentially regulates critically important pathways for follicular oocyte maturation, changed in follicular fluid EVs [110]. Extracellular vesicles isolated from bovine follicular fluid contain miRNAs that reflect the stage of the estrus cycle and can modulate cumulus cell transcription during in vitro maturation [111]. Murine oEVs can transfer miR-34c-5p to the sperm heads where it localizes to the centromere and promotes the first cleavage in the zygote [112]. In addition, oEVs carry miRNAs that potentially target several embryonic development related genes [112]. Oviductal EVs were observed to fuse with canine oocytes and release miRNAs with a potential role in oocyte maturation and follicular growth [113]. In vitro, oEV supplementation altered the bovine transcriptome, suggesting a possible role of oEV miRNA cargo in controlling embryonic development [114]. Extracellular vesicles secreted by donor oviductal cells increased birth rates after embryo transfer in mice due to decreased apoptosis and improved cellular differentiation in embryos [115]. A miRNA mediated embryo-maternal communication was also reported and Cuman et al. (2015) [116], demonstrated that human embryos secrete miR-661 that was up-taken by endometrial epithelial cells and inhibited cell adhesiveness [116]. To note, miR-661 exchange was observed to be mediated by the RNA binding complex (RBC) protein Argonaute 1 rather than EV shuttling [99], but it is likely that other EV encapsulated miRNAs can potentially be shuttled from embryos to maternal tissue. Recently, the presence of microRNAs and extracellular vesicles in human blastocoel fluid was reported, suggesting a possible role of EV miRNA cargo in regulation of blastocyst development. MiRNA expression in blastocoel fluid reflects the miRNome of embryonic cells with abundant miRNA targeting genes regulating critical signaling pathways controlling embryo development [117]. Syncytial-like cells, derived from murine trophoblast stem cells, secrete EVs with a miRNA transcript from the X chromosome cluster miR-322-3p and 5p, miR-503-5p, miR-542-3p, miR-450a-5p, miR-292-3p and mmu-miR-183-5p that was found to be increased in EVs isolated from the sera of pregnant mice [118]. In vitro and in vivo studies have reported miRNA EVs shuttling from endometrium to embryos. During the window of implantation, human endometrium epithelium releases EVs carrying specific miRNAs [8]. Among them, miR-30d was observed to be encapsulated in EVs and released by primary human endometrial epithelial cells (hEECs) and transferred to the trophectoderm of murine embryos when co-cultured, thus regulating genes related to embryonic adhesion and promoting embryo implantation [8]. MicroR-34c-5p was found to be differentially enriched in EVs isolated from mouse endometrium during different phases of pregnancy, and it is likely to target GAS1 (Growth Arrest Specific 1) and influence embryo implantation [119]. In sheep, a clear influence of progesterone-mediated regulation of miRNA content in EVs isolated from the uterine lumen was reported [120]. An RNA-based communication occurred between uterus and embryo also through functional mRNA-encapsulating EVs. In vitro study of co-culture of human trophoblast spheroids and human endometrial cell lines showed that EVs package and transport specific RNAs (intronic and exonic LINC00478, and ZNF81) from trophoblast to endometrial cells, that results in a reduced level of the corresponding transcripts in the endometrial receiving cell [121]). Finally, Simon et al. [122] first characterized the DNA-cargo of the EVs by the embryo showing a random representation of embryo genomic DNA inconsistent with aneuploidy or apoptotic cellular events [122].

7. Extracellular Vesicles as Biomarkers in Reproductive Medicine

The EVs are important vectors of information and are involved both in physiological and pathological conditions. Indeed, during diseases, cell metabolism, physiology and behavior are modified causing alteration in cellular gene expression, protein content and EV cargo. This makes EVs important in the scientific field for their functional role in cell communication and in the medical field as biomarker or as therapeutic tools.
The physiological level of circulating EVs in tissues, serum or other biological fluids is different considering the animal model, the time (hours or days) after disease, the kind of disease and the EV quantification method. However, this information, studied and identified in the different conditions, could help to identify the possible therapeutic dose.
In the reproductive tract, EV-mediated communication between cells and embryo–maternal crosstalk is an essential process that occurs both in physiologic and in pathologic conditions. Therefore, molecular cargo of EVs and its release change in the pathologic condition by affecting maintenance, and progression of reproductive- and obstetric-related pathologies (Table 3).

7.1. Biomarkers for Female Reproductive Cancer

Extracellular vesicles can regulate a variety of physiologic processes. The molecular cargo of EVs is strictly dependent on the status of the cell or tissue donor and can change in pathologic conditions such as cardiac disease, neurologic diseases and cancer [132]. Although, tissue biopsies are the gold standard for cancer evaluation, they are invasive and provide only a small sample of the whole tumour. Moreover, it can be difficult to collect sequential samples to monitor the progression of the tumour with time and response to therapy [133].
Extracellular vesicles eliminate these limitations. They are present in all biological fluids (including urine, blood, saliva and cerebral spinal fluid), and their involvement in different diseases make them inviting candidates as biomarkers. Collection of body fluids for screening of some diseases could avoid the necessity for invasive examinations [134]. Inside the EVs, the molecular cargo is protected from degradation and can be used as biomarkers for non-invasive cancer diagnosis [135,136]. For example, specific extracellular vesicle-derived miRNAs are associated with eight types of cancer (including lung, breast, and ovarian) [137]. In addition, analysis of miRNA from some EVs may be used to distinguish between benign and malignant disease. Indeed, expression levels of miR-200 family members (miR-200a, miR-200b and miR-200c) in one study could distinguish malignant from benign ovarian tumours with a sensitivity of 88% and specificity of 90% [123]. The strength of miRNAs as ovarian tumour markers is confirmed by the fact that elevated miR-200b and miR-200c levels are correlated with serum CA125 levels which is a current standard of diagnosis, monitoring treatment reaction and predicting prognosis [123].

7.2. Biomarkers for Female Fertility

Physiologic alterations occurring in the reproductive tract in pathologic conditions influence cell metabolism, changing gene expression and protein content and thus altering EV concentration, cargo and function. These alterations influence maintenance and progression of reproductive- and obstetric-related pathologies.
In human medicine, decrease in fertility is an important issue. This may be correlated to environmental stressors. Phthalates and phenols are classes of potential endocrine disrupting chemicals present mainly in the environment and in food [138]. Phthalates and phenols have been shown to influence fertility in women [139]. A correlation has been identified between the concentration of phthalates and phenols and their impact on the EV-miRNA profile in follicular fluid with a negative impact on female fertility [124].
Several of the significant EV-miRNAs biomarkers found to be associated with phthalates and phenols play key roles in the ovarian follicle, consistent with the fact that the follicular fluid, contains a number of molecules including EV-miRNAs that create an appropriate microenvironment for the growth of oocytes [140,141]. For example, increased urinary concentration of phenols has been associated with altered expression of miR-125b, miR-24 and miR-375, which have been shown to play an important role in animal oocyte maturation and fertilization. In rodents, over expression of a miR-125b mimic block expression of specific genes required for embryos to progress beyond the two-cell stage [142]. Higher levels of miR-24 in bovine culture media is associated with embryos failing to undergo differentiation [143]. MicroR-375 is expressed in granulosa cells and oocytes and targets genes that regulate follicular growth proliferation, spread and apoptosis of cumulus cells [132,133]. Overexpression of miR-375 blocked the ability to proliferate, increased the apoptosis rate of cumulus cells in cows [144,145] and suppressed estradiol production and follicular development in porcine granulosa cells [146].
These data show that exposure to toxic compounds causes miRNAs alteration in human EVs isolated from follicular fluid, potentially affecting ovarian function. In the veterinary field, it is likely that exposure to toxic compounds such as Fusarium mycotoxins, frequently present in feed, might influence animal fertility and alter the miRNA expression in follicular fluid EVs.
In man, but probably also in animals, fertility status is influenced by other factors such as advanced maternal age (AMA) or endometriosis that can impact the embryo-endometrial talk before implantation. In vitro studies revealed variation of EV-bound secreted miRNAs isolated from co-cultured endometrial cells and embryos from AMA and fertile controls, with 16 miRNAs increase in AMA and strong evidence of a negative regulation toward 206 target genes involved in cell adhesion. Co-cultured endometrial cells isolated from patients with endometriosis and embryos from fertile controls revealed EV alterations in 10 miRNAs, targeting 1014 genes crucial for development and implantation [125].

7.3. Biomarkers for Embryo Quality

Currently, in vitro embryo production is an important component of improved pregnancy rates in assisted reproduction in both human and veterinary medicine. Blastocyst quality is assessed morphologically by assessment of the expansion of the blastocoel cavity, as well as the appearance of the ICM and TE cells [147,148,149,150]. Another important technique is removal of cells from the TE by biopsy [151]. In human medicine, this method is used to detect, and thus prevent, transmission of single gene disorders such as cystic fibrosis and β-thalassemia [152]. The identification of specific markers for embryo quality, would avoid the need for embryo biopsy and its inherent risks. In 2013, for the first time, genomic DNA of unknown origin was identified in blastocoel fluid (BF) and in embryo culture medium [153]. However, the use of mitochondrial DNA to assess embryo quality is not used because of the presence of other contaminant DNA present in the culture media from maternal (cumulus) cells [154].
The miRNAs found outside the blastocyst in the spent medium are probably involved in implantation and in embryo-maternal cross-talking [116,155,156,157]. Since embryos secrete miRNAs in culture medium in vitro and in the uterus in vivo, it is conceivable that the extracellular vesicles, carrying miRNAs, DNA and other molecules, might reflect embryo quality [85]. Indeed, some authors show that the expression profile of miRNAs reflects embryo aneuploidies and that the miRNAs can be used to evaluate embryo implantation potential [140]. Extracellular vesicle cargo released in spent culture medium depends on the physiologic state of the embryo and reflects the in vitro embryo quality before implantation or its cultural environment. For example, during in vitro embryo culture, EV concentration in low oxygen tension increased on Day 3 and decreased on Day 7 and miR-210 levels can be used as a marker for normoxia since it is associated with low oxygen tension [158].A method (based on characterizing EVs by flow cytometry), using propidium iodide for DNA staining, showed that low numbers of stained vesicles is associated with embryo competence indicating reduced cell injury and embryo damage and thus less degraded DNA in the EVs [126]. MicroRNA profiling of spent blastocyst culture media from implanted versus unplanted blastocysts showed increased expression of specific miRNA in degenerate conditioned media in bovine [138] samples, probably associated with different EV cargo. However, Cimadomo et al. [159] reported that medium from poor-quality blastocysts contained more miRNAs probably as a result of passive miRNA accumulation in the medium because of cellular turnover rather than active secretion from EVs.

7.4. Biomarkers for Placenta Quality

Pregnancy is a particular physiological condition for studying EVs. At first, unlike other conditions, the onset and end of pregnancy is known allowing you to follow changes in EVs throughout the period. In addition, the placenta produces specific markers that allow you to distinguish EVs from those produced by others cell types and, finally, the placenta is available at the end of pregnancy for study [160]. Placental secreted EVs are important in feto-maternal communication and have been identified in maternal blood. Placental EVs were isolated from maternal blood by a chromatographic/immunosorbent procedure using antibody agarose beads against placental alkaline phosphatase (PLAP), a protein present exclusively on EVs secreted from placentae [161]. Placenta-derived EVs may be differentiated from other EVs by the presence of placenta specific proteins (eg, PLAP4) and miRNAs (eg, chromosome 19 miRNA cluster) that are exclusively expressed in the placenta [53,161,162].
During gestation, placenta-derived EVs can be bio-markers of feto–maternal health and evolution, indeed, EVs are involved in cell-to-cell communication between the placenta and peripheral blood immune cells. During the first trimester of pregnancy, extravillous trophoblast and/or syncytiotrophoblast cells release placental EVs into the maternal blood. These EVs suppress maternal T-cell signaling components promoting immunosuppression and maternal immune tolerance to the fetus. In man, the plasma concentration of EVs increases during pregnancy and with gestational age, indeed, the concentration of EVs in maternal peripheral blood is 20-fold greater compared to that found in nonpregnant women [125]. The increased level of EVs is also found in several pathologic conditions [163] and for this reason changes in exosome levels may be of clinical utility in the diagnosis of placental dysfunction. Indeed, it has been demonstrated that some proteins (for example the specific syncytiotrophoblast protein, syncytin-2) are markedly down-regulated in EVs derived from the placentae of pregnant women with preeclampsia compared to healthy control pregnancies [127]. For this reason, placental EVs and specific miRNAs have been further examined as biomarkers for diagnosis of pre-eclampsia [128]. Plasma EVs isolated from pre-eclampsia pregnancies were shown to impair angiogenesis of human umbilical vein endothelial cells and to expressed abundant sFlt-1 (soluble fms-like tyrosine kinase-1) and sEng (soluble endoglin) [102].
In addition, the secretion of EVs from the placenta can be influenced by the microenvironment. Indeed, variation in oxygen tension and glucose can increase bioactivity and release of exosome by trophoblast [98,164]. Extracellular vesicles isolated from placenta of pregnant women with diabetes mellitus show alterations in the protein content related to energy production, metabolism and inflammation [129] and alteration in miRNA associated with skeletal muscle insulin sensitivity [130].
Sometime, the effects of high glucose concentration on the bioactivity of EVs are additive to those of oxygen tension when there is an association with maternal insulin resistance (resulting in hyperglycemia) and pre-eclampsia (associated with placental insufficiency and hypoxia) [165]. The identification of such biomarkers would be useful screening tests to identify those asymptomatic women who develop common pathologies [165].

7.5. Biomarker for Early Abortion

Sabapatha et al. (2006) evaluating the content of miRNAs in pregnant and control women report that the plasma concentration of EVs increases during pregnancy and with advancing gestational age [161]. Pohler et al. (2017) found 194 and 211 circulating extracellular vesicle-derived miRNAs from Days 17 and 24 of gestation confirming their increase during gestation [131]. However, three miRNAs (miR-25, -16a/b, and -3596) were present in greater abundance either in controls or in cows suffering embryonic-mortality compared to pregnant animals on Day 17. The increase in miR-25 abundance in cows with embryonic-mortality might indicate early embryonic death or a systemic response to pregnancy loss. Pathway analysis indicated that the changes to specific miRNAs in cows with embryonic-mortality versus pregnant animals could affect the up-regulation of pathways involved in prostaglandin production [166]. It is known that prostaglandin is responsible for corpus luteum regression and that with this involution the concentrations of progesterone, critical for pregnancy establishment, fall [167,168]. In bovine pregnant female blood, some miRNAs can be down or up-regulates in circulation exosomes demonstrating the potential role of circulating exosomal miRNAs as biomarkers in early embryonic mortality or early pregnancy diagnosis [169]. It would be interesting to validate differentially abundant miRNAs to determine biomarkers for the diagnosis of early pregnancy success or embryonic mortality [131].

8. EVs and Therapeutic Action

Since endometrial exosomes are implicated in the pathophysiology of implantation failure in infertility [7], current research is attempting to exploit the clinical utility of EVs. Extracellular vesicles are easy to manipulate because they are composed of biological membranes that protect their contents. Small molecules, proteins or RNA, anti-cancer agents, anti-inflammatory compounds, miRNA, mRNA, proteins, and growth factors can be loaded into EVs via transfection or electroporation [170,171]. EVs can be loaded using endogenous or exogenous approaches. The endogenous method first loads therapeutics into parent cells, followed by the generation and release of loaded EVs [172]. In the exogenous approach, therapeutic agents are incorporated into isolated EVs through an incubation process [173,174].
The EVs can be used for treatment of cancer and can be loaded with anti-cancer agents (in cells or in EVs) including doxorubicin, paclitaxel, curcumin, cisplatin, and methotrexate [175]. Extracellular vesicles have had enormous therapeutic potential in neurologic disorders [176], and show regenerative effects in animal models of neurological, cardiovascular, liver, kidney, and skin diseases [177]. In addition, their ability to promote re-epithelialization of skin wounds and stroke recovery has been reported [178]. Recently, amniotic derived EVs were used to treat endometritis in mare with the goal of achieving successful pregnancy [179]. This mare was an 11-year-old Friesian, with a history of failed pregnancies despite numerous insemination attempts for many years. After a final insemination attempt using a stallion of proven fertility, the collection of an eight-day old embryo suggested that the mare was affected by implantation failure related to endometritis. The regenerative medicine using amniotic derived microvesicles was a new approach for this disease. The mare was treated with two cycles of intrauterine administration of amniotic-derived microvesicles that induced an improvement in the classification of endometritis and a pregnancy ended with the birth of the foal. This is the only paper the used EVs in reproductive disease. Probably, amniotic microvesicles for their anti-inflammatory and regenerative effects [52] were able to restore the injured endometrium and re-establish the proper communication for a successful embryo implantation.
Extracellular vesicles can be administered via intravenous, subcutaneous, intraperitoneal, oral, and intranasal routes to reach the desired in vivo target sites, depending on the intended therapeutic action and tissues to be targeted [180]. In the paper by Lange-Consiglio et al. [179] EVs were administered by the intravaginal rout. The biodistribution and targeting potential of EVs is dependent on EV sizes, surface markers, and their isolation methods. Limitations influencing EV biodistribution through various routes of administration include transport through several specific physical barriers, non-target tissue uptake, immune cell-dependent phagocytosis, and rapid in vivo clearance [180].
There are many considerations for optimization of the therapeutic action of EVs and to ensure treatments are safe. These include: 1) the cell type (such as the tissue source of MSCs); 2) the cell collection process and the expansion methods including culture conditions (growth medium and supplement used, type of bioreactor, number of passages performed, etc.); 3) the EV isolation and storage methods; 4) the modality of drug-loading and the quantification approaches; 5) the mechanism used to trigger EV release; 6) the design of cost-effective and scalable isolation processes and 7) the methods to ensure quality, safety, and consistency that are mandatory for clinical applications [180]. Variations in these parameters can influence the EV population size, generation (yield/cell), membrane markers, EV contents including miRNAs, the purity and safety profile of EVs [181] and conceivably their therapeutic action too.

9. Conclusions

This review summarizes the role of EVs in cell to cell communication in animal reproduction. Remarkable progress has already been made, but many hurdles remain. Although, there is clearly evidence that EVs play crucial roles in many aspects of oocyte maturation, fertilization, implantation and embryo-maternal crosstalk, challenges remain in a partial understanding of which signaling biomolecules carried by EVs act to mediate cell to cell communication. In fact, cells release a heterogeneous population of EVs differing in size and molecular content that are often co-purified using conventional isolation methods. Molecular profiling gives an overall characterization of EV cargo that only partially represents the function of specific EV subpopulations in physiologic and pathologic conditions. The development of more reliable isolation methods and more sensitive technologies for physical and molecular characterization of EVs is needed to obtain more robust results that will improve our insight into the molecular basis of EV targeting of, and action on, recipient cells. Moreover, a more precise vision of the cocktail of molecules inside EVs that are transferred in the two-way trafficking between embryos and mothers will improve the identification of potential non-invasive biomarkers for specific pathologic conditions and will advance the use of EVs in innovative therapies in reproductive biomedicine.

Author Contributions

A.L.-C. and E.C. wrote the manuscript. E.C. produced Figures and Tables. Both co-authors provided useful comments on the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ng, Y.H.; Rome, S.; Jalabert, A.; Forterre, A.; Singh, H.; Hincks, C.L.; Salamonsen, L.A. Endometrial exosomes/microvesicles in the uterine microenvironment: A new paradigm for embryo-endometrial cross talk at implantation. PLoS ONE 2013, 8, e58502. [Google Scholar] [CrossRef] [Green Version]
  2. Xu, R.; Greening, D.W.; Zhu, H.J.; Takahashi, N.; Simpson, R.J. Extracellular vesicle isolation and characterization: Toward clinical application. J. Clin. Invest. 2016, 126, 1152–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. 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]
  4. Andronico, F.; Battaglia, R.; Ragusa, M.; Barbagallo, D.; Purrello, M.; Di Pietro, C. Extracellular vesicles in human oogenesis and implantation. Int. J. Mol. Sci. 2019, 20, 2162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Almiñana, C.; Bauersachs, S. Extracellular vesicles: Multi-signal messengers in the gametes/embryo-oviduct cross-talk. Theriogenology 2020, 150, 59–69. [Google Scholar] [CrossRef] [PubMed]
  6. Bridi, A.; Perecin, F.; Silveira, J.C.D. Extracellular vesicles mediated early embryo-maternal interactions. Int. J. Mol. Sci. 2020, 21, 1163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Gurung, S.; Greening, D.W.; Catt, S.; Salamonsen, L.; Evans, J. Exosomes and soluble secretome from hormone-treated endometrial epithelial cells direct embryo implantation. Mol. Hum. Reprod. 2020, 26, 510–520. [Google Scholar] [CrossRef] [PubMed]
  8. Vilella, F.; Moreno-Moya, J.M.; Balaguer, N.; Grasso, A.; Herrero, M.; Martinez, S.; Marcilla, A.; Simon, C. Hsa-miR-30d, secreted by the human endometrium, is taken up by the pre-implantation embryo and might modify its transcriptome. Development 2015, 142, 3210–3221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Balaguer, N.; Moreno, I.; Herrero, M.; González, M.; Simón, C.; Vilella, F. Heterogeneous nuclear ribonucleoprotein C1 may control miR-30d levels in endometrial exosomes affecting early embryo implantation. Mol. Hum. Reprod. 2018, 24, 411–425. [Google Scholar] [CrossRef] [PubMed]
  10. Greening, D.W.; Nguyen, H.P.; Elgass, K.; Simpson, R.J.; Salamonsen, L.A. Human endometrial exosomes contain hormone-specific cargo modulating trophoblast adhesive capacity: Insights into endometrial-embryo interactions. Biol. Reprod. 2016, 94, 38. [Google Scholar] [CrossRef]
  11. Evans, J.; Rai, A.; Nguyen, H.P.T.; Poh, Q.H.; Elglass, K.; Simpson, R.J.; Salamonsen, L.A.; Greening, D.W. Human endometrial extracellular vesicles functionally prepare human trophectoderm model for implantation: Understanding bidirectional maternal-embryo communication. Proteomics 2019, 19, e1800423. [Google Scholar] [CrossRef]
  12. Simon, C.; Greening, D.W.; Bolumar, D.; Balaguer, N.; Salamonsen, L.A.; Vilella, F. Extracellular vesicles in human reproduction in health and disease. Endocr. Rev. 2018, 39, 292–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Burnett, L.A.; Nowak, R.A. Exosomes mediate embryo and maternal interactions at implantation and during pregnancy. Front. Biosci. (Schol Ed.) 2016, 8, 79–96. [Google Scholar] [CrossRef] [Green Version]
  14. Crawford, N. The presence of contractile proteins in platelet microparticles isolated from human and animal platelet-free plasma. Br. J. Haematol. 1971, 21, 53–69. [Google Scholar] [CrossRef] [PubMed]
  15. Stegmayr, B.; Ronquist, G. Promotive effect on human sperm progressive motility by prostasomes. Urol. Res. 1982, 10, 253–257. [Google Scholar] [CrossRef]
  16. Trams, E.G.; Lauter, C.J.; Salem, N., Jr.; Heine, U. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim. Biophys. Acta 1981, 645, 63–70. [Google Scholar] [CrossRef]
  17. Johnstone, R.M.; Hammond, J.R.; Turbide, C. Vesicle formation during reticulocyte maturation. association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar]
  18. Makarova, K.S.; Yutin, N.; Bell, S.D.; Koonin, E.V. Evolution of diverse cell division and vesicle formation systems in Archaea. Nat. Rev. Microbiol. 2010, 8, 731–741. [Google Scholar] [CrossRef] [Green Version]
  19. Kim, J.H.; Lee, J.; Park, J.; Gho, Y.S. Gram-negative and Gram-positive bacterial extracellular vesicles. Semin. Cell Dev. Biol. 2015, 40, 97–104. [Google Scholar] [CrossRef]
  20. Oliveira, D.L.; Nakayasu, E.S.; Joffe, L.S.; Guimarães, A.J.; Sobreira, T.J.; Nosanchuk, J.D.; Cordero, R.J.; Frases, S.; Casadevall, A.; Almeida, I.C.; et al. Characterization of yeast extracellular vesicles: Evidence for the participation of different pathways of cellular traffic in vesicle biogenesis. PLoS ONE 2010, 5, e11113. [Google Scholar] [CrossRef] [Green Version]
  21. Harding, C.; Heuser, J.; Stahl, P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J. Cell Biol. 1983, 97, 329–339. [Google Scholar] [CrossRef]
  22. 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 Biol. 1985, 101, 942–948. [Google Scholar] [CrossRef] [Green Version]
  23. McConnell, R.E.; Higginbotham, J.N.; Shifrin, D.A., Jr; Tabb, D.L.; Coffey, R.J.; Tyska, M.J. The enterocyte microvillus is a vesicle-generating organelle. J. Cell Biol. 2009, 185, 1285–1298. [Google Scholar] [CrossRef] [Green Version]
  24. Muralidharan-Chari, V.; Clancy, J.; Plou, C.; Romao, M.; Chavrier, P.; Raposo, G.; D’Souza-Schorey, C. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr. Biol. 2009, 19, 1875–1885. [Google Scholar] [CrossRef] [Green Version]
  25. Konoshenko, M.Y.; Lekchnov, E.A.; Vlassov, A.V.; Laktionov, P.P. Isolation of extracellular vesicles: General methodologies and latest trends. Biomed. Res. Int. 2018, 2018, 8545347. [Google Scholar] [CrossRef]
  26. Böing, A.N.; van der Pol, E.; Grootemaat, A.E.; Coumans, F.A.; Sturk, A.; Nieuwland, R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles 2014, 3, 23430. [Google Scholar] [CrossRef] [PubMed]
  27. Deregibus, M.C.; Figliolini, F.; D’Antico, S.; Manzini, P.M.; Pasquino, C.; De Lena, M.; Tetta, C.; Brizzi, M.F.; Camussi, G. Charge-based precipitation of extracellular vesicles. Int. J. Mol. Med. 2016, 38, 1359–1366. [Google Scholar] [CrossRef] [Green Version]
  28. Kenigsberg, S.; Wyse, B.A.; Librach, C.L.; da Silveira, J. Protocol for exosome isolation from small volume of ovarian follicular fluid: Evaluation of ultracentrifugation and commercial kits. Methods Mol. Biol. 2017, 1660, 321–341. [Google Scholar] [CrossRef]
  29. Brennan, K.; Martin, K.; FitzGerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci. Rep. 2020, 10, 1039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Nakai, W.; Yoshida, T.; Diez, D.; Miyatake, Y.; Nishibu, T.; Imawaka, N.; Naruse, K.; Sadamura, Y.; Hanayama, R. A novel affinity-based method for the isolation of highly purified extracellular vesicles. Sci. Rep. 2016, 6, 33935. [Google Scholar] [CrossRef] [Green Version]
  31. Sidhom, K.; Obi, P.O.; Saleem, A. A Review of exosomal isolation methods: Is size exclusion chromatography the best option? Int. J. Mol. Sci. 2020, 21, 6466. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, H.; Lyden, D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat. Protoc. 2019, 14, 1027–1053. [Google Scholar] [CrossRef]
  33. Zhang, Q.; Higginbotham, J.N.; Jeppesen, D.K.; Yang, Y.P.; Li, W.; McKinley, E.T.; Graves-Deal, R.; Ping, J.; Britain, C.M.; Dorsett, K.A.; et al. Transfer of functional cargo in exomeres. Cell Rep. 2019, 27, 940–954. [Google Scholar] [CrossRef] [Green Version]
  34. Witwer, K.W.; Buzás, E.I.; Bemis, L.T.; Bora, A.; Lässer, C.; Lötvall, J.; Nolte-’t Hoen, E.N.; Piper, M.G.; Sivaraman, S.; Skog, J.; et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicles 2013, 2. [Google Scholar] [CrossRef]
  35. 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 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Wu, Y.; Deng, W.; Klinke, D.J., 2nd. Exosomes: Improved methods to characterize their morphology, RNA content, and surface protein biomarkers. Nanoparticle Tracking Analysis (NTA) is commonly used to determine EV concentration and diameter. Analyst 2015, 140, 6631–6642. [Google Scholar] [CrossRef] [Green Version]
  37. Cizmar, P.; Yuana, Y. Detection and characterization of extracellular vesicles by transmission and cryo-transmission electron microscopy. Methods Mol. Biol. 2017, 1660, 221–232. [Google Scholar] [CrossRef] [PubMed]
  38. Szatanek, R.; Baj-Krzyworzeka, M.; Zimoch, J.; Lekka, M.; Siedlar, M.; Baran, J. The Methods of choice for extracellular vesicles (EVs) characterization. Int. J. Mol. Sci. 2017, 18, 1153. [Google Scholar] [CrossRef] [PubMed]
  39. Kim, A.; Ng, W.B.; Bernt, W.; Cho, N.J. Validation of size estimation of nanoparticle tracking analysis on polydisperse macromolecule assembly. Sci. Rep. 2019, 9, 2639. [Google Scholar] [CrossRef] [Green Version]
  40. Noble, J.M.; Roberts, L.M.; Vidavsky, N.; Chiou, A.E.; Fischbach, C.; Paszek, M.J.; Estroff, L.A.; Kourkoutis, L.F. Direct comparison of optical and electron microscopy methods for structural characterization of extracellular vesicles. J. Struct. Biol. 2020, 210, 107474. [Google Scholar] [CrossRef]
  41. Menck, K.; Bleckmann, A.; Schulz, M.; Ries, L.; Binder, C. Isolation and characterization of microvesicles from peripheral blood. J. Vis. Exp. 2017, 119, 55057. [Google Scholar] [CrossRef] [Green Version]
  42. Shao, H.; Im, H.; Castro, C.M.; Breakefield, X.; Weissleder, R.; Lee, H. New technologies for analysis of extracellular vesicles. Chem. Rev. 2018, 118, 1917–1950. [Google Scholar] [CrossRef]
  43. Greening, D.W.; Simpson, R.J. Understanding extracellular vesicle diversity - current status. Expert Rev. Proteom. 2018, 15, 887–910. [Google Scholar] [CrossRef]
  44. Claridge, B.; Kastaniegaard, K.; Stensballe, A.; Greening, D.W. Post-translational and transcriptional dynamics—Regulating extracellular vesicle biology. Expert Rev. Proteom. 2019, 16, 17–31. [Google Scholar] [CrossRef]
  45. Kreimer, S.; Belov, A.M.; Ghiran, I.; Murthy, S.K.; Frank, D.A.; Ivanov, A.R. Mass-spectrometry-based molecular characterization of extracellular vesicles: Lipidomics and proteomics. J. Proteome Res. 2015, 14, 2367–2384. [Google Scholar] [CrossRef]
  46. Haraszti, R.A.; Didiot, M.C.; Sapp, E.; Leszyk, J.; Shaffer, S.A.; Rockwell, H.E.; Gao, F.; Narain, N.R.; DiFiglia, M.; Kiebish, M.A.; et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J. Extracell. Vesicles 2016, 5, 32570. [Google Scholar] [CrossRef]
  47. Record, M.; Silvente-Poirot, S.; Poirot, M.; Wakelam, M.J.O. Extracellular vesicles: Lipids as key components of their biogenesis and functions. J. Lipid Res. 2018, 59, 1316–1324. [Google Scholar] [CrossRef] [Green Version]
  48. Vagner, T.; Chin, A.; Mariscal, J.; Bannykh, S.; Engman, D.M.; Di Vizio, D. Protein composition reflects extracellular vesicle heterogeneity. Proteomics 2019, 19, e1800167. [Google Scholar] [CrossRef] [PubMed]
  49. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  50. Desrochers, L.M.; Bordeleau, F.; Reinhart-King, C.A.; Cerione, R.A.; Antonyak, M.A. Microvesicles provide a mechanism for intercellular communication by embryonic stem cells during embryo implantation. Nat. Commun. 2016, 7, 11958. [Google Scholar] [CrossRef] [Green Version]
  51. Collino, F.; Deregibus, M.C.; Bruno, S.; Sterpone, L.; Aghemo, G.; Viltono, L.; Tetta, C.; Camussi, G. Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs. PLoS ONE 2010, 5, e11803. [Google Scholar] [CrossRef]
  52. Lange-Consiglio, A.; Lazzari, B.; Perrini, C.; Pizzi, F.; Stella, A.; Cremonesi, F.; Capra, E. MicroRNAs of Equine amniotic mesenchymal cell-derived microvesicles and their involvement in anti-inflammatory processes. Cell Transpl. 2018, 27, 45–54. [Google Scholar] [CrossRef] [Green Version]
  53. Donker, R.B.; Mouillet, J.F.; Chu, T.; Hubel, C.A.; Stolz, D.B.; Morelli, A.E.; Sadovsky, Y. The expression profile of C19MC microRNAs in primary human trophoblast cells and exosomes. Mol. Hum. Reprod. 2012, 18, 417–424. [Google Scholar] [CrossRef] [Green Version]
  54. Tosar, J.P.; Gambaro, F.; Sanguinetti, J.; Bonilla, B.; Witwer, K.W.; Cayota, A. Assessment of small RNA sorting into different extracellular fractions revealed by high-throughput sequencing of breast cell lines. Nucleic Acids Res. 2015, 43, 5601–5616. [Google Scholar] [CrossRef] [Green Version]
  55. Mateescu, B.; Kowal, E.J.; van Balkom, B.W.; Bartel, S.; Bhattacharyya, S.N.; Buzás, E.I.; Buck, A.H.; de Candia, P.; Chow, F.W.; Das, S.; et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA—An ISEV position paper. J. Extracell. Vesicles 2017, 6, 1286095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chevillet, J.R.; Kang, Q.; Ruf, I.K.; Briggs, H.A.; Vojtech, L.N.; Hughes, S.M.; Cheng, H.H.; Arroyo, J.D.; Meredith, E.K.; Gallichotte, E.N.; et al. Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proc. Natl. Acad. Sci. USA 2014, 111, 14888–14893. [Google Scholar] [CrossRef] [Green Version]
  57. Lange-Consiglio, A.; Lazzari, B.; Pizzi, F.; Stella, A.; Girani, A.; Quintè, A.; Cremonesi, F.; Capra, E. different culture times affect microRNA cargo in equine amniotic mesenchymal cells and their microvesicles. Tissue Eng, Part C Methods 2018, 24, 596–604. [Google Scholar] [CrossRef]
  58. Willms, E.; Cabañas, C.; Mäger, I.; Wood, M.J.A.; Vader, P. Extracellular vesicle heterogeneity: Subpopulations.; isolation techniques.; and diverse functions in cancer progression. Front. Immunol. 2018, 9, 738. [Google Scholar] [CrossRef] [Green Version]
  59. Margolis, L.; Sadovsky, Y. The biology of extracellular vesicles: The known unknowns. PLoS Biol. 2019, 17, e3000363. [Google Scholar] [CrossRef]
  60. Buzás, E.I.; Tóth, E.Á.; Sódar, B.W.; Szabó-Taylor, K.É. Molecular interactions at the surface of extracellular vesicles. Semin. Immunopathol. 2018, 40, 453–464. [Google Scholar] [CrossRef] [Green Version]
  61. Wu, D.; Yan, J.; Shen, X.; Sun, Y.; Thulin, M.; Cai, Y.; Wik, L.; Shen, Q.; Oelrich, J.; Qian, X.; et al. Profiling surface proteins on individual exosomes using a proximity barcoding assay. Nat. Commun. 2019, 10, 3854. [Google Scholar] [CrossRef]
  62. Mulcahy, L.A.; Pink, R.C.; Carter, D.R. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3. [Google Scholar] [CrossRef] [Green Version]
  63. Horibe, S.; Tanahashi, T.; Kawauchi, S.; Murakami, Y.; Rikitake, Y. Mechanism of recipient cell-dependent differences in exosome uptake. BMC Cancer 2018, 18, 47. [Google Scholar] [CrossRef] [Green Version]
  64. Kanada, M.; Bachmann, M.H.; Hardy, J.W.; Frimannson, D.O.; Bronsart, L.; Wang, A.; Sylvester, M.D.; Schmidt, T.L.; Kaspar, R.L.; Butte, M.J.; et al. Differential fates of biomolecules delivered to target cells via extracellular vesicles. Proc. Natl. Acad. Sci. USA 2015, 112, E1433–E1442. [Google Scholar] [CrossRef] [Green Version]
  65. Ono, R.; Yasuhiko, Y.; Aisaki, K.I.; Kitajima, S.; Kanno, J.; Hirabayashi, Y. Exosome-mediated horizontal gene transfer occurs in double-strand break repair during genome editing. Commun. Biol. 2019, 2, 57. [Google Scholar] [CrossRef]
  66. 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. Biol. Chem. 2014, 289, 3869–3875. [Google Scholar] [CrossRef] [Green Version]
  67. 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]
  68. Bai, R.; Latifi, Z.; Kusama, K.; Nakamura, K.; Shimada, M.; Imakawa, K. Induction of immune-related gene expression by seminal exosomes in the porcine endometrium. Biochem. Biophys. Res. Commun. 2018, 495, 1094–1101. [Google Scholar] [CrossRef]
  69. Al-Dossary, A.A.; Strehler, E.E.; Martin-Deleon, P.A. Expression and secretion of plasma membrane Ca2þ-ATPase 4a (PMCA4a) during murine estrus: Association with oviductal exosomes and uptake in sperm. PLoS ONE 2013, 8, e80181. [Google Scholar] [CrossRef] [Green Version]
  70. Bathala, P.; Fereshteh, Z.; Li, K.; Al-Dossary, A.A.; Galileo, D.S.; Martin-DeLeon, P.A. Oviductal extracellular vesicles (oviductosomes, OVS) are conserved in humans: Murine OVS play a pivotal role in sperm capacitation and fertility. Mol. Hum. Reprod. 2018, 24, 143–157. [Google Scholar] [CrossRef]
  71. Ferraz, M.d.A.M.M.; Carothers, A.; Dahal, R.; Noonan, M.J.; Songsasen, N. Oviductal extracellular vesicles interact with the spermatozoon’s head and mid-piece and improves its motility and fertilizing ability in the domestic cat. Sci. Rep. 2019, 9, 9484. [Google Scholar] [CrossRef]
  72. Franchi, A.; Moreno-Irusta, A.; Domínguez, E.M.; Adre, A.J.; Giojalas, L.C. Extracellular vesicles from oviductal isthmus and ampulla stimulate the induced acrosome reaction and signaling events associated with capacitation in bovine spermatozoa. J. Cell Biochem. 2020, 121, 2877–2888. [Google Scholar] [CrossRef]
  73. Almiñana, C.; Corbin, E.; Tsikis, G.; Alcântara-Neto, A.S.; Labas, V.; Reynaud, K.; Galio, L.; Uzbekov, R.; Garanina, A.S.; Druart, X.; et al. Oviduct extracellular vesicles protein content and their role during oviduct–embryo cross-talk. Reproduction 2017, 154, 253–268. [Google Scholar] [CrossRef]
  74. Lopera-Vasquez, R.; Hamdi, M.; Maillo, V.; Gutierrez-Adan, A.; Bermejo-Alvarez, P.; Ramírez, M.Á.; Yáñez-Mó, M.; Rizos, D. Effect of bovine oviductal extracellular vesicles on embryo development and quality in vitro. Reproduction 2017, 153, 461–470. [Google Scholar] [CrossRef] [Green Version]
  75. da Silveira, J.C.; Andrade, G.M.; Del Collado, M.; Sampaio, R.V.; Sangalli, J.R.; Silval, L.A.; Pinaffi, F.V.L.; Jardim, I.B.; Cesar, M.C.; Coutinho, L.L.; et al. Supplementation with small-extracellular vesicles from ovarian follicular fluid during in vitro production modulates bovine embryo development. PLoS ONE 2017, 12, e0179451. [Google Scholar] [CrossRef]
  76. Bidarimath, M.; Khalaj, K.; Kridli, R.T.; Kan, F.W.K.; Koti, M.; Tayade, C. Extracellular vesicle mediated intercellular communication at the porcine maternal-fetal interface: A new paradigm for conceptus-endometrial cross-talk. Sci. Rep. 2017, 7, 40476. [Google Scholar] [CrossRef] [PubMed]
  77. Nakamura, K.; Kusama, K.; Bai, R.; Sakurai, T.; Isuzugawa, K.; Godkin, J.D.; Suda, Y.; Imakawa, K. Induction of IFNT-stimulated genes by conceptus-derived exosomes during the attachment period. PLoS ONE 2016, 11, e0158278. [Google Scholar] [CrossRef] [Green Version]
  78. Salomon, C.; Kobayashi, M.; Ashman, K.; Sobrevia, L.; Murray, M.D.; Rice, G.E. Hypoxia-induced changes in the bioactivity of cytotrophoblast-derived exosomes. PLoS ONE 2013, 8, e79636. [Google Scholar] [CrossRef] [Green Version]
  79. Kusama, K.; Nakamura, K.; Bai, R.; Nagaoka, K.; Sakurai, T.; Imakawa, K. Intrauterine exosomes are required for bovine conceptus implantation. Biochem. Biophys. Res. Commun. 2018, 495, 1370–1375. [Google Scholar] [CrossRef]
  80. Wang, X.; Tian, F.; Chen, C.; Feng, Y.; Sheng, X.; Guo, Y.; Ni, H. Exosome-derived uterine microRNAs isolated from cows with endometritis impede blastocyst development. Reprod. Biol. 2019, 19, 204–209. [Google Scholar] [CrossRef] [PubMed]
  81. Riou, C.; Brionne, A.; Cordeiro, L.; Harichaux, G.; Gargaros, A.; Labas, V.; Gautron, J.; Gérard, N. Avian uterine fluid proteome: Exosomes and biological processes potentially involved in sperm survival. Mol. Reprod. Dev. 2020, 87, 454–470. [Google Scholar] [CrossRef]
  82. Pons-Rejraji, H.; Artonne, C.; Sion, B.; Brugnon, F.; Canis, M.; Janny, L.; Grizard, G. Prostasomes: Inhibitors of capacitation and modulators of cellular signaling in human sperm. Int. J. Androl. 2011, 34, 568–580. [Google Scholar] [CrossRef]
  83. Piehl, L.L.; Fischman, M.L.; Hellman, U.; Cisale, H.; Miranda, P.V. Boar seminal plasma exosomes: Effect on sperm function and protein identification by sequencing. Theriogenology 2013, 79, 1071–1082. [Google Scholar] [CrossRef] [PubMed]
  84. Da Silveira, J.C.; Veeramachaneni, D.N.R.; Winger, Q.A.; Carnevale, E.M.; Bouma, G.J. Cell-secreted vesicles in equine ovarian follicular fluid contain mirnas and proteins: A possible new form of cell communication within the ovarian follicle. Biol. Reprod 2012, 86, 71. [Google Scholar] [CrossRef] [PubMed]
  85. Sang, Q.; Yao, Z.; Wang, H.; Feng, R.; Wang, H.; Zhao, X.; Xing, Q.; Jin, L.; He, L.; Wu, L.; et al. Identification of microRNAs in human follicular fluid: Characterization of microRNAs that govern steroidogenesis in vitro and are associated with polycystic ovary syndrome in vivo. J. Clin. Endocrinol. Metab. 2013, 98, 3068–3079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Andrade, G.; Meirelles, F.; Perecin, F.; da Silveira, J. Cellular and extracellular vesicular origins of miRNAs within the bovine ovarian follicle. Reprod. Domest. Anim. 2017, 52, 1036–1045. [Google Scholar] [CrossRef]
  87. Spitschak, M.; Hoeflich, A. Potential functions of IGFBP-2 for ovarian folliculogenesis and steroidogenesis. Front. Endocrinol. 2018, 9, 119. [Google Scholar] [CrossRef] [Green Version]
  88. Bourdiec, A.; Akoum, A. Embryo implantation: Role of interleukin 1 family members. Med. Sci. (Paris) 2014, 30, 644–650. [Google Scholar] [CrossRef] [Green Version]
  89. Bourdiec, A.; Shao, R.; Rao, C.V.; Akoum, A. Human chorionic gonadotropin triggers angiogenesis via the modulation of endometrial stromal cell responsiveness to interleukin 1: A new possible mechanism underlying embryo implantation. Biol. Reprod. 2012, 87, 6. [Google Scholar] [CrossRef]
  90. Stavreus-Evers, A.; Koraen, L.; Scott, J.E.; Zhang, P.; Westlund, P. Distribution of cyclooxygenase-1, cyclooxygenase-2, and cytosolic phospholipase A2 in the luteal phase human endometrium and ovary. Fertil. Steril. 2005, 83, 156–162. [Google Scholar] [CrossRef]
  91. Jessmon, P.; Leach, R.E.; Armant, D.R. Diverse functions of HBEGF during pregnancy. Mol. Reprod. Dev. 2009, 76, 1116–1127. [Google Scholar] [CrossRef] [Green Version]
  92. Wang, H.; Shi, G.; Li, M.; Fan, H.; Ma, H.; Sheng, L. Correlation of IL-1 and HB-EGF with endometrial receptivity. Exp. Ther. Med. 2018, 16, 5130–5136. [Google Scholar] [CrossRef] [Green Version]
  93. Merviel, P.; Challier, J.C.; Carbillon, L.; Foidart, J.M.; Uzan, S. The role of integrins in human embryo implantation. Fetal Diagn, Ther. 2001, 16, 364–371. [Google Scholar] [CrossRef]
  94. Salleh, N.; Giribabu, N. Leukemia inhibitory factor: Roles in embryo implantation and in nonhormonal contraception. Sci. World J. 2014, 2014, 201514. [Google Scholar] [CrossRef] [Green Version]
  95. Mardpour, S.; Hamidieh, A.A.; Taleahmad, S.; Sharifzad, F.; Taghikhani, A.; Baharvand, H. Interaction between mesenchymal stromal cell-derived extracellular vesicles and immune cells by distinct protein content. J. Cell Physiol. 2019, 234, 8249. [Google Scholar] [CrossRef]
  96. Burns, G.W.; Brooks, K.E.; Spencer, T.E. Extracellular vesicles originate from the conceptus and uterus during early pregnancy in sheep. Biol. Reprod. 2016, 94, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kshirsagar, S.K.; Alam, S.M.; Jasti, S.; Hodes, H.; Nauser, T.; Gilliam, M.; Billstrand, C.; Hunt, J.S.; Petroff, M.G. Immunomodulatory molecules are released from the first trimester and term placenta via exosomes. Placenta 2012, 33, 982–990. [Google Scholar] [CrossRef] [Green Version]
  98. Mincheva-Nilsson, L.; Baranov, V. Placenta-derived exosomes and syncytiotrophoblast microparticles and their role in human reproduction: Immune modulation for pregnancy success. Am. J. Reprod Immunol. 2014, 72, 440–457. [Google Scholar] [CrossRef]
  99. Giacomini, E.; Alleva, E.; Fornelli, G.; Quartucci, A.; Privitera, L.; Vanni, V.S.; Viganò, P. Embryonic extracellular vesicles as informers to the immune cells at the maternal-fetal interface. Clin. Exp. Immunol. 2019, 198, 15–23. [Google Scholar] [CrossRef] [Green Version]
  100. Lokossou, A.G.; Toudic, C.; Nguyen, P.T.; Elissee, X.; Vargas, A.; Rassart, É.; Lafond, J.; Leduc, L.; Bourgault, S.; Gilbert, C.; et al. Endogenous retrovirus-encoded syncytin-2 contributes to exosome-mediated immunosuppression of t cells. Biol. Reprod. 2020, 102, 185–198. [Google Scholar] [CrossRef]
  101. Pillay, P.; Moodley, K.; Moodley, J.; Mackraj, I. Placenta-derived exosomes: Potential biomarkers of preeclampsia. Int. J. Nanomed. 2017, 12, 8009–8023. [Google Scholar] [CrossRef] [Green Version]
  102. Chang, X.; Yao, J.; He, Q.; Liu, M.; Duan, T.; Wang, K. Exosomes from women with preeclampsia induced vascular dysfunction by delivering sFlt (Soluble Fms-Like Tyrosine Kinase)-1 and sEng (Soluble Endoglin) to endothelial cells. Hypertension 2018, 72, 1381–1390. [Google Scholar] [CrossRef]
  103. Gézsi, A.; Kovács, Á.; Visnovitz, T.; Buzás, E.I. Systems Biology approaches to investigating the roles of extracellular vesicles in human diseases. Exp. Mol. Med. 2019, 51, 1–11. [Google Scholar] [CrossRef] [Green Version]
  104. Burkova, 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]
  105. Tong, M.; Kleffmann, T.; Pradhan, S.; Johansson, C.L.; DeSousa, J.; Stone, P.R.; James, J.L.; Chen, Q.; Chamley, L.W. Proteomic characterization of macro-, micro- and nano-extracellular vesicles derived from the same first trimester placenta: Relevance for feto-maternal communication. Hum. Reprod. 2016, 31, 687–699. [Google Scholar] [CrossRef]
  106. Almiñana, C.; Tsikis, G.; Labas, V.; Uzbekov, R.; da Silveira, J.C.; Bauersachs, S.; Mermillod, P. Deciphering the oviductal extracellular vesicles content across the estrous cycle: Implications for the gametes-oviduct interactions and the environment of the potential embryo. BMC Genom. 2018, 19, 622. [Google Scholar] [CrossRef] [Green Version]
  107. Griffiths, G.S.; Reese, K.L.; Galileo, D.S.; Martin-DeLeon, P.A. Investigating the role of murine epididymosomes and uterosomes in GPI-linked protein transfer to sperm using SPAM1 as a model. Mol. Reprod. Dev. 2008, 75, 1627–1636. [Google Scholar] [CrossRef]
  108. Szekeres-Bartho, J.; Šućurović, S.; Mulac-Jeričević, B. The Role of extracellular vesicles and PIBF in embryo-maternal immune-interactions. Front. Immunol. 2018, 9, 2890. [Google Scholar] [CrossRef]
  109. Herrera-Van Oostdam, A.S.; Salgado-Bustamante, M.; López, J.A.; Herrera-Van Oostdam, D.A.; López-Hernández, Y. Placental exosomes viewed from an ’omics’ perspective: Implications for gestational diabetes biomarkers identification. Biomark. Med. 2019, 13, 675–684. [Google Scholar] [CrossRef]
  110. Santonocito, M.; Vento, M.; Guglielmino, M.R.; Battaglia, R.; Wahlgren, J.; Ragusa, M.; Barbagallo, D.; Borzì, P.; Rizzari, S.; Maugeri, M.; et al. Molecular characterization of exosomes and their microRNA cargo in human follicular fluid: Bioinformatic analysis reveals that exosomal microRNAs control pathways involved in follicular maturation. Fertil. Steril. 2014, 102, 1751–1761. [Google Scholar] [CrossRef]
  111. de Ávila, A.C.F.C.M.; Bridi, A.; Andrade, G.M.; Del Collado, M.; Sangalli, J.R.; Nociti, R.P.; da Silva Junior, W.A.; Bastien, A.; Robert, C.; Meirelles, F.V.; et al. Estrous cycle impacts microRNA content in extracellular vesicles that modulate bovine cumulus cell transcripts during in vitro maturation. Biol. Reprod. 2020, 102, 362–375. [Google Scholar] [CrossRef]
  112. Fereshteh, Z.; Schmidt, S.A.; Al-Dossary, A.A.; Accerbi, M.; Arighi, C.; Cowart, J.; Song, J.L.; Green, P.J.; Choi, K.; Yoo, S.; et al. Murine Oviductosomes (OVS) microRNA profiling during the estrous cycle: Delivery of OVS-borne micro-RNAs to sperm where miR-34c-5p localizes at the centrosome. Sci. Rep. 2018, 8, 16094. [Google Scholar] [CrossRef]
  113. Lange-Consiglio, A.; Perrini, C.; Albini, G.; Modina, S.; Lodde, V.; Orsini, E.; Esposti, P.; Cremonesi, F. Oviductal microvesicles and their effect on in vitro maturation of canine oocytes. Reproduction 2017, 154, 167–180. [Google Scholar] [CrossRef]
  114. Bauersachs, S.; Mermillod, P.; Almiñana, C. The oviductal extracellular vesicles RNA cargo regulates the bovine embryonic transcriptome. Int. J. Mol. Sci. 2020, 21, 1303. [Google Scholar] [CrossRef] [Green Version]
  115. Qu, P.; Zhao, Y.; Wang, R.; Zhang, Y.; Li, L.; Fan, J.; Liu, E. Extracellular vesicles derived from donor oviduct fluid improved birth rates after embryo transfer in mice. Reprod. Fertil. Dev. 2019, 31, 324–332. [Google Scholar] [CrossRef] [Green Version]
  116. Cuman, C.; Van Sinderen, M.; Gantier, M.P.; Rainczuk, K.; Sorby, K.; Rombauts, L.; Osianlis, T.; Dimitriadis, E. Human blastocyst secreted microRNA regulate endometrial epithelial cell adhesion. EBioMedicine 2015, 2, 1528–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Battaglia, R.; Palini, S.; Vento, M.E.; La Ferlita, A.; Lo Faro, M.J.; Caroppo, E.; Borzì, P.; Falzone, L.; Barbagallo, D.; Ragusa, M.; et al. Identification of extracellular vesicles and characterization of miRNA expression profiles in human blastocoel fluid. Sci. Rep. 2019, 9, 84. [Google Scholar] [CrossRef]
  118. Stefanski, A.L.; Martinez, N.; Peterson, L.K.; Callahan, T.J.; Treacy, E.; Luck, M.; Friend, S.F.; Hermesch, A.; Maltepe, E.; Phang, T.; et al. Murine trophoblast-derived and pregnancy-associated exosome-enriched extracellular vesicle microRNAs: Implications for placenta driven effects on maternal physiology. PLoS ONE 2019, 14, e0210675. [Google Scholar] [CrossRef] [Green Version]
  119. Tan, Q.; Shi, S.; Liang, J.; Zhang, X.; Cao, D.; Wang, Z. MicroRNAs in small extracellular vesicles indicate successful embryo implantation during early pregnancy. Cells 2020, 9, 645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Burns, G.W.; Brooks, K.E.; O’Neil, E.V.; Hagen, D.E.; Behura, S.K.; Spencer, T.E. Progesterone effects on extracellular vesicles in the sheep uterus. Biol. Reprod. 2018, 98, 612–622. [Google Scholar] [CrossRef]
  121. Es-Haghi, M.; Godakumara, K.; Häling, A.; Lättekivi, F.; Lavrits, A.; Viil, J.; Andronowska, A.; Nafee, T.; James, V.; Jaakma, Ü.; et al. Specific trophoblast transcripts transferred by extracellular vesicles affect gene expression in endometrial epithelial cells and may have a role in embryo-maternal crosstalk. Cell Commun. Signal. 2019, 17, 146. [Google Scholar] [CrossRef] [Green Version]
  122. Simon, B.; Bolumar, D.; Amadoz, A.; Jimenez-Almazán, J.; Valbuena, D.; Vilella, F.; Moreno, I. Identification and characterization of extracellular vesicles and its DNA cargo secreted during murine embryo development. Genes 2020, 11, 203. [Google Scholar] [CrossRef] [Green Version]
  123. Meng, X.; Muller, V.; Milde-Langosch, K.; Trillsch, F.; Pantel, K.; Schwarzenbach, H. Diagnostic and prognostic relevance of circulating exosomal miR-373, miR-200a, miR-200b and miR-200c in patients with epithelial ovarian cancer. Oncotarget 2016, 7, 16923–16935. [Google Scholar] [CrossRef] [Green Version]
  124. Martinez, R.M.; Hauser, R.; Liang, L.; Mansur, A.M.; Dioni, L.; Racowsky, C.; Bollati, V.; Baccarelli, A.A.; Machtinger, R. Urinary concentrations of phenols and phthalate metabolites reflect extracellular vesicle microRNA expression in follicular fluid. Environ. Int. 2019, 123, 20–28. [Google Scholar] [CrossRef]
  125. Parks, J.C.; McCallie, B.R.; Patton, A.L.; Al-Safi, Z.A.; Polotsky, A.J.; Griffin, D.K.; Schoolcraft, W.B.; Katz-Jaffe, M.G. The impact of infertility diagnosis on embryo-endometrial dialogue. Reproduction 2018, 155, 543–552. [Google Scholar] [CrossRef]
  126. Pallinger, E.; Bognar, Z.; Bodis, J.; Csabai, T.; Farkas, N.; Godony, K.; Varnagy, A.; Buzas, E.; Szekeres-Bartho, J. A simple and rapid flow cytometry-based assay to identify a competent embryo prior to embryo transfer. Sci. Rep. 2017, 7, 39927. [Google Scholar] [CrossRef]
  127. Vargas, A.; Zhou, S.; Ethier-Chiasson, M.; Flipo, D.; Lafond, J.; Gilbert, C.; Barbeau, B. Syncytin proteins incorporated in placenta exosomes are important for cell uptake and show variation in abundance in serum exosomes from patients with preeclampsia. FASEB J. 2014, 28, 3703–3719. [Google Scholar] [CrossRef]
  128. Li, H.; Ouyang, Y.; Sadovsky, E.; Parks, W.T.; Chu, T.; Sadowsky, Y. Unique microRNA signals in plasma exosomes from pregnancies complicated by preeclampsia. Hypertension 2020, 75, 762–771. [Google Scholar] [CrossRef]
  129. Jayabalan, N.; Lai, A.; Nair, S.; Guanzon, D.; Scholz-Romero, K.; Palma, C.; McIntyre, H.D.; Lappas, M.; Salomon, C. Quantitative proteomics by SWATH-MS suggest an association between circulating exosomes and maternal metabolic changes in gestational diabetes mellitus. Proteomics 2019, 19, e1800164. [Google Scholar] [CrossRef] [Green Version]
  130. Nair, S.; Jayabalan, N.; Guanzon, D.; Palma, C.; Scholz-Romero, K.; Elfeky, O.; Zuñiga, F.; Ormazabal, V.; Diaz, E.; Rice, G.E.; et al. Human placental exosomes in gestational diabetes mellitus carry a specific set of miRNAs associated with skeletal muscle insulin sensitivity. Clin. Sci. (Lond) 2018, 132, 2451–2467. [Google Scholar] [CrossRef]
  131. Pohler, K.G.; Green, J.A.; Moley, L.A.; Gunewardena, S.; Hung, W.T.; Payton, R.R.; Hong, X.; Christenson, L.K.; Geary, T.W.; Smith, M.F. Circulating microRNA as candidates for early embryonic viability in cattle. Mol. Reprod. Dev. 2017, 84, 731–743. [Google Scholar] [CrossRef]
  132. Larrea, E.; Sole, C.; Manterola, L.; Goicoechea, I.; Armesto, M.; Arestin, M.; Caffarel, M.M.; Araujo, A.M.; Araiz, M.; Fernandez-Mercado, M.; et al. New concepts in cancer biomarkers: Circulating miRNAs in liquid biopsies. Int. J. Mol. Sci. 2016, 17, 627. [Google Scholar] [CrossRef] [Green Version]
  133. Barger, J.F.; Rahman, M.A.; Jackson, D.; Acunzo, M.; Nana-Sinkam, S.P. Extracellular miRNAs as biomarkers in cancer. Food Chem. Toxicol. 2016, 98, 66–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Lin, J.; Li, J.; Huang, B.; Liu, J.; Chen, X.; Chen, X.M.; Xu, Y.M.; Huang, L.F.; Wang, X.Z. Exosomes: Novel biomarkers for clinical diagnosis. Sci. World J. 2015, 2015, 657086. [Google Scholar] [CrossRef]
  135. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable bloodbased markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Chen, I.H.; Aguilar, H.A.; PaezPaez, J.S.; Wu, X.; Pan, L.; Wendt, M.K.; Iliuk, A.B.; Zhang, Y.; Tao, W.A. Analytical pipeline for discovery and verification of glycoproteins from plasma-derived extracellular vesicles as breast cancer biomarkers. Anal. Chem. 2018, 90, 6307–6313. [Google Scholar] [CrossRef]
  137. Thind, A.; Wilson, C. Exosomal miRNAs as cancer biomarkers and therapeutic targets. J. Extracell. Vesicles 2016, 5, 31292. [Google Scholar] [CrossRef]
  138. Diamanti-Kandarakis, E.; Bourguignon, J.P.; Giudice, L.C.; Hauser, R.; Prins, G.S.; Soto, A.M.; Zoeller, R.T.; Gore, A.C. Endocrine-disrupting chemicals: An endocrine society scientific statement. Endocr. Rev. 2009, 30, 293–342. [Google Scholar] [CrossRef]
  139. Machtinger, R.; Gaskins, A.J.; Racowsky, C.; Mansur, A.; Adir, M.; Baccarelli, A.A.; Calafat, A.M.; Hauser, R. Urinary concentrations of biomarkers of phthalates and phthalate alternatives and IVF outcomes. Environ. Int. 2018, 111, 23–31. [Google Scholar] [CrossRef]
  140. Rodgers, R.J.; Irving-Rodgers, H.F. Formation of the ovarian follicular antrum and follicular fluid. Biol. Reprod. 2010, 82, 1021–1029. [Google Scholar] [CrossRef] [Green Version]
  141. Zuccotti, M.; Merico, V.; Cecconi, S.; Redi, C.A.; Garagna, S. What does it take to make a developmentally competent mammalian egg? Hum. Reprod. Update 2011, 17, 525–540. [Google Scholar] [CrossRef] [Green Version]
  142. Kim, K.H.; Seo, Y.M.; Kim, E.Y.; Lee, S.Y.; Kwon, J.; Ko, J.J.; Lee, K.A. The miR-125 family is an important regulator of the expression and maintenance of maternal effect genes during preimplantational embryo development. Open Biol. 2016, 6, 160181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Kropp, J.; Khatib, H. Characterization of microRNA in bovine in vitro culture media associated with embryo quality and development. J. Dairy Sci. 2015, 98, 6552–6563. [Google Scholar] [CrossRef] [Green Version]
  144. Chen, H.; Liu, C.; Jiang, H.; Gao, Y.; Xu, M.; Wang, J.; Liu, S.; Fu, Y.; Sun, X.; Xu, J.; et al. Regulatory role of miRNA-375 in expression of BMP15/GDF9 receptors and its effect on proliferation and apoptosis of bovine cumulus cells. Cell. Physiol. Biochem. 2017, 41, 439–450. [Google Scholar] [CrossRef]
  145. Liu, C.; Yuan, B.; Chen, H.; Xu, M.; Sun, X.; Xu, J.; Gao, Y.; Chen, C.; Jiang, H.; Zhang, J. Effects of MiR-375-BMPR2 as a key factor downstream of BMP15/GDF9 on the Smad1/5/8 and Smad2/3 signaling pathways. Cell. Physiol. Biochem. 2018, 46, 213–225. [Google Scholar] [CrossRef] [Green Version]
  146. Yu, C.; Li, M.; Wang, Y.; Liu, Y.; Yan, C.; Pan, J.; Liu, J.; Cui, S. miR-375 mediates CRH signaling pathway in inhibiting E2 synthesis in porcine ovary. Reproduction 2017, 153, 63–73. [Google Scholar] [CrossRef] [Green Version]
  147. International Embryo Transfer Society. Manual of the international embryo transfer society. In IETS Manual; Stringfellow, D.A., Seidel, S.M., Eds.; The Society: Savoy, IL, USA, 1998. [Google Scholar]
  148. Gardner, D.K.; Schoolcraf, W.B. Culture and transfer of human blastocysts. Curr. Opin. Obstet. Gynecol. 1999, 11, 307–311. [Google Scholar] [CrossRef]
  149. Veeck, L.L.; Bodine, R.; Clarke, R.N.; Berrios, R.; Libraro, J.; Moschini, R.M.; Zaninovic, N.; Rosenwaks, Z. High pregnancy rates can be achieved after freezing and thawing human blastocysts. Fertil. Steril. 2004, 82, 1418–1427. [Google Scholar] [CrossRef]
  150. Stephenson, E.L.; Braude, P.R.; Mason, C. International community consensus standard for reporting derivation of humanembryonic stem cell lines. Regen. Med. 2007, 2, 349–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Scott, R.T.; Upham, K.M.; Forman, E.J.; Zhao, T.; Treff, N.R. Cleavage-stage biopsy significantly impairs human embryonic implantation potential while blastocyst biopsy does not: A randomized and paired clinical trial. Fertil. Steril. 2013, 100, 624–630. [Google Scholar] [CrossRef]
  152. Gutiérrez-Mateo, C.; Sánchez-García, J.F.; Fischer, J.; Tormasi, S.; Cohen, J.; Munné, S.; Wells, D. Preimplantation genetic diagnosis of single-gene disorders: Experience with more than 200 cycles conducted by a reference laboratory in the United States. Fertil. Steril. 2009, 92, 1544–1556. [Google Scholar] [CrossRef]
  153. Palini, S.; Galluzzi, L.; De Stefani, S.; Bianchi, M.; Wells, D.; Magnani, M.; Bulletti, C. Genomic DNA in human blastocoelefluid. Reprod. Biomed. Online 2013, 26, 603–610. [Google Scholar] [CrossRef] [Green Version]
  154. Hammond, E.R.; McGillivray, B.C.; Wicker, S.M.; Peek, J.C.; Shelling, A.N.; Stone, P.; Chamley, L.W.; Cree, L.M. Characterizing nuclear and mitochondrial DNA in spent embryo culture media: Genetic contamination identified. Fertil. Steril. 2017, 107, 220–228. [Google Scholar] [CrossRef] [Green Version]
  155. Rosenbluth, E.M.; Shelton, D.N.; Wells, L.M.; Sparks, A.E.; Van Voorhis, B.J. Human embryos secrete microRNAs into culture media–a potential biomarker for implantation. Fertil. Steril. 2014, 101, 1493–1500. [Google Scholar] [CrossRef]
  156. Galliano, D.; Pellicer, A. MicroRNA and implantation. Fertil. Steril. 2014, 101, 1531–1544. [Google Scholar] [CrossRef] [PubMed]
  157. Giacomini, E.; Vago, R.; Sanchez, A.M.; Podini, P.; Zarovni, N.; Murdica, V.; Rizzo, R.; Bortolotti, D.; Candiani, M.; Viganò, P. Secretome of in vitro cultured human embryos contains extracellular vesicles that are uptaken by the maternal side. Sci. Rep. 2017, 7, 5210. [Google Scholar] [CrossRef]
  158. Andrade, G.M.; Bomfim, M.M.; del Collado, M.; Meirelles, F.V.; Perecin, F.; da Silveira, J.C. Oxygen tension modulates extracellular vesicles and its miRNA contents in bovine embryo culture medium. Mol. Reprod. Dev. 2019, 86, 1067–1080. [Google Scholar] [CrossRef]
  159. Cimadomo, D.; Rienzi, L.; Giancani, A.; Alviggi, E.; Dusi, L.; Canipari, R.; Noli, L.; Ilic, D.; Khalaf, Y.; Ubaldi, F.M.; et al. Definition and validation of a custom protocol to detect miRNAs in the spent media after blastocyst culture: Searching for biomarkers of implantation. Hum. Reprod. 2019, 34, 1746–1761. [Google Scholar] [CrossRef]
  160. Tannetta, D.; Dragovic, R.; Alyahyaei, Z.; Southcombe, J. Extracellular vesicles and reproduction–promotion of successful pregnancy. Cell. Mol. Immunol. 2014, 11, 548–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Sabapatha, A.; Gercel-Taylor, C.; Taylor, D.D. Specific isolation of placenta-derived exosomes from the circulation of pregnant women and their immunoregulatory consequences. Am. J. Reprod. Immunol. 2006, 56, 345–355. [Google Scholar] [CrossRef]
  162. Ouyang, Y.; Mouillet, J.F.; Coyne, C.B.; Sadovsky, Y. Review: Placenta-specific micro- RNAs in exosomesegood things come in nanopackages. Placenta 2014, 35, S69–S73. [Google Scholar] [CrossRef] [Green Version]
  163. Pap, E.; Pallinger, E.; Falus, A.; Kiss, A.A.; Kittel, A.; Kovács, P.; Buzás, E.I. T lymphocytes are targets for platelet- and trophoblast-derived microvesicles during pregnancy. Placenta 2008, 29, 826–832. [Google Scholar] [CrossRef]
  164. Salomon, C.; Scholz-Romero, K.; Kobayashi, M.; Smith, M.; Duncombe, M.; Hanes, G.; Mitchell, S.; Rice, M.D.; Gregory, E. Oxygen tension regulates glucose-induced biogenesis and release of different subpopulations of exosome vesicles from trophoblast cells: A gestational age profile of placental exosomes in maternal plasma with gestational diabetes mellitus. Placenta 2015, 36, 488. [Google Scholar] [CrossRef]
  165. Mitchell, M.D.; Peiris, H.N.; Kobayashi, M.; Koh, Y.Q.; Duncombe, G.; Illanes, S.E.; Rice, G.E.; Salomon, C. Placental exosomes in normal and complicated pregnancy. Am. J. Obstet. Gynecol. 2015, S173–S181. [Google Scholar] [CrossRef]
  166. Bazer, F.W. Pregnancy recognition signaling mechanisms in ruminants and pigs. J. Anim. Sci. Biotechnol. 2013, 4, 1. [Google Scholar] [CrossRef] [Green Version]
  167. Godkin, J.; Bazer, F.; Thatcher, W.; Roberts, R. Proteins released by cultured day 15-16 conceptuses prolong luteal maintenance when introduced into the uterine lumen of cyclic ewes. J. Reprod. Fertil. 1984, 71, 57–64. [Google Scholar] [CrossRef] [Green Version]
  168. Roberts, R.; Imakawa, K.; Niwano, Y.; Kazemi, M.; Malathy, P.-V.; Hansen, T.; Glass, A.A.; Kronenberg, L. Interferon production by the preimplantation sheep embryo. J. Interferon Res. 1989, 9, 175–187. [Google Scholar] [CrossRef]
  169. De Bem, T.H.C.; da Silveira, J.C.; Sampaio, R.V.; Sangalli, J.R.; Oliveira, M.L.F.; Ferreira, R.M.; Silva, L.A.; Perecin, F.; King, W.A.; Meirelles, F.V.; et al. Low levels of exosomal-miRNAs in maternal blood are associated with early pregnancy loss in cloned cattle. Sci. Rep. 2017, 7, 14319. [Google Scholar] [CrossRef]
  170. Andaloussi, S.E.L.; Mager, I.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
  171. Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio Rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef]
  172. Pascucci, L.; Coccè, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Viganò, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Control. Release 2014, 192, 262–270. [Google Scholar] [CrossRef]
  173. Tian, Y.; Li, S.; Song, J.; Ji, T.; Zhu, M.; Anderson, G.J.; Wei, J.; Nie, G. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014, 35, 2383–2390. [Google Scholar] [CrossRef] [PubMed]
  174. Yang, J.; Wei, F.; Schafer, C.; Wong, D.T.W. Detection of tumor cell-specific mRNA and protein in exosome-like microvesicles from blood and saliva. PLoS ONE 2014, 9. [Google Scholar] [CrossRef]
  175. Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular vesicles in cancer - implications for future improvements in cancer care. Nat. Rev. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef]
  176. Vinaiphat, A.; Sze, S.K. Clinical implications of extracellular vesicles in neurodegenerative diseases. Expert Rev. Mol. Diagn. 2019, 1–12. [Google Scholar] [CrossRef]
  177. Lelek, J.; Zuba-Surma, E.K. Perspectives for future use of extracellular vesicles from umbilical cord- and adipose tissue-derived mesenchymal stem/stromal cells in regenerative therapies-synthetic review. Int. J. Mol. Sci. 2020, 21, 799. [Google Scholar] [CrossRef] [Green Version]
  178. Riazifar, M.; Pone, E.J.; Lötvall, J.; Zhao, W. Stem cell extracellular vesicles: Extended messages of regeneration. Annu. Rev. Pharmacol. Toxicol. 2017, 57, 125–154. [Google Scholar] [CrossRef] [Green Version]
  179. Lange-Consiglio, A.; Funghi, F.; Cantile, C.; Idda, A.; Cremonesi, F.; Riccaboni, P. Case report: Use of amniotic microvesicles for regenerative medicine treatment of a mare with chronic endometritis. Front. Vet. Sci. 2020, 7, 347. [Google Scholar] [CrossRef]
  180. Burnouf, T.; Agrahari, V.; Agrahari, V. Extracellular vesicles as nanomedicine: Hopes and hurdles in clinical translation. Int. J. Nanomed. 2019, 14, 8847–8859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Gimona, M.; Pachler, K.; Laner-Plamberger, S.; Schallmoser, K.; Rohde, E. Manufacturing of human extracellular vesicle-based therapeutics for clinical use. Int. J. Mol Sci. 2017, 18, 1190. [Google Scholar] [CrossRef]
Figure 1. Overview of the main procedures used for the isolation, characterization and clinical use of extracellular vesicles (EVs). (1) Extracellular vesicles are released by cells into the culture media or from tissue into the extracellular environment. (2) Extracellular vesicles are separated or further purified to obtain a more homogeneous EV population using a variety of methods. (3) Isolated EVs are physically characterized by: Scanning Electron Microscopes (SEM), Transmission Electron Microscopes (TEM), Nanoparticle Tracking Analysis (NTA) and Dynamic Light Scattering (DLS). (4) Extracellular vesicles are molecularly characterized using several techniques. (5) Biomarkers can be obtained by molecular profiling of isolated EVs from in vitro cell cultures or body fluids in pathologic condition and used as diagnostic tools for several human and veterinary diseases. Isolated EVs can also be used as treatments in human and veterinary medicine.
Figure 1. Overview of the main procedures used for the isolation, characterization and clinical use of extracellular vesicles (EVs). (1) Extracellular vesicles are released by cells into the culture media or from tissue into the extracellular environment. (2) Extracellular vesicles are separated or further purified to obtain a more homogeneous EV population using a variety of methods. (3) Isolated EVs are physically characterized by: Scanning Electron Microscopes (SEM), Transmission Electron Microscopes (TEM), Nanoparticle Tracking Analysis (NTA) and Dynamic Light Scattering (DLS). (4) Extracellular vesicles are molecularly characterized using several techniques. (5) Biomarkers can be obtained by molecular profiling of isolated EVs from in vitro cell cultures or body fluids in pathologic condition and used as diagnostic tools for several human and veterinary diseases. Isolated EVs can also be used as treatments in human and veterinary medicine.
Biomolecules 10 01510 g001
Figure 2. Graphical representation of the EVs mediated cross-talk interaction in the female reproductive system. (1) Seminal plasma EVs (spEVs) interact with endometrium (2) oviductal tract EVs (oEVs)with sperm (2a) and embryo (2b),(3) follicular fluids EVs (ffEVs) with embryo (4) embryo EVs (eEVs) with endometrium (5) placenta EVs (pEVs)with extravillous trophoblasts (6) endometrium EVs (endEVs) with embryo. Colored dashed line (and numbers) indicates EVs release and uptake from donor and recipient cells or tissue, which studies are reported in Table 2. In figure, the human female reproductive tract was shown as example of mammalian female reproductive system.
Figure 2. Graphical representation of the EVs mediated cross-talk interaction in the female reproductive system. (1) Seminal plasma EVs (spEVs) interact with endometrium (2) oviductal tract EVs (oEVs)with sperm (2a) and embryo (2b),(3) follicular fluids EVs (ffEVs) with embryo (4) embryo EVs (eEVs) with endometrium (5) placenta EVs (pEVs)with extravillous trophoblasts (6) endometrium EVs (endEVs) with embryo. Colored dashed line (and numbers) indicates EVs release and uptake from donor and recipient cells or tissue, which studies are reported in Table 2. In figure, the human female reproductive tract was shown as example of mammalian female reproductive system.
Biomolecules 10 01510 g002
Table 1. Summary of EV isolation techniques and main advantages (pros) and disadvantages (cons) for different methods.
Table 1. Summary of EV isolation techniques and main advantages (pros) and disadvantages (cons) for different methods.
MethodPrincipleAdvantagesDisadvantages
Differential centrifugationEVs isolation after different consequent centrifugation steps (from 300g to 100,000 g): depletion of cells, platelets and large apoptotic bodies by low-speed centrifugation steps. Larger EVs are pelleted at 10,000 20,000 g range. Smaller EVs are then pelleted at high speed (100,000 120,000 g).Most used, intermediated recovery and specificityTime consuming and extravesicular proteins complexes/aggregates, lipoprotein particles, and other contaminants may also sediment
Density gradient centrifugationEVs isolation through density gradients of sucrose or iohexol or iodixanol High purity (EVs float upward or move downwards into an overlaid density gradient)Applicable to small volume (usually after differential centrifugation), sucrose or iohexol or iodixanol can influence downstream application
FiltrationEVs filtration with different molecular weight cutoff Recovery and purity depend on the consequent centrifugation step and the cutoff of centrifugal filter employLow specificity, EV populations may adhere to the filter or filtering may cause deformation and breakup of large vesicles
PrecipitationEVs are precipitated with organic solvent or in presence of different chemical compound such as polyethylene glycol (PEG), sodium acetate or protamineHigh recovery, fast and easyLow specificity Coprecipitation of numerous non-EV contaminants such as lipoproteins. Rigorous assessment of contaminating particle is recommended
Size Exclusion Chromatography EVs are separated by their ability to pass through a resin packed in a columnWell separated EVs from protein complexes biofluidsNot suitable for large volume
Affinity isolationEVs bind specific antibodies against exosome-specific surface proteins or EV-binding molecules High purity Low recovery, requires specific antibody
Microfluidic devicesMicrofluidics-based on-chip EVs isolation based on immunoaffinity, membrane filtration, nanowire-based traps, nano-sized deterministic lateral displacement, viscoelastic flow and acoustic isolation high-throughput and low processing timeNot suitable for large volume
Nanoscale flow cytometric sortingFluorescent labelled EVs are sorted using flow cytometerVery specific and high purityLaborious and time consuming
Table 2. Extracellular vesicles mediated cross-talk between different compartments of the reproductive system as shown in Figure 2. Extracellular vesicles released from different cell or body fluids (seminal plasma, oviduct, follicular fluid, embryo, placental mesenchymal stem cells, endometrial epithelial cells) and from different species (bos taurus, felis catus, homo sapiens, mus musculus, ovis aries, sus scrofa), target different cell types. Method of isolation, main results and references for each EV-mediated exchange are also shown.
Table 2. Extracellular vesicles mediated cross-talk between different compartments of the reproductive system as shown in Figure 2. Extracellular vesicles released from different cell or body fluids (seminal plasma, oviduct, follicular fluid, embryo, placental mesenchymal stem cells, endometrial epithelial cells) and from different species (bos taurus, felis catus, homo sapiens, mus musculus, ovis aries, sus scrofa), target different cell types. Method of isolation, main results and references for each EV-mediated exchange are also shown.
Ref. Figure 2EVs Isolated fromSpeciesIsolation MethodTarget CellPhysical CharacterizationMain ResultsReferences
1seminal plasmasus scrofapolymer precipitationendometrial epithelial cells TEM, NTAinduction of immune-related gene expression in endometrial epithelial cells EECs[68]
2aoviductmus musculusultracentrifugationSpermTEMPMCA4 sperm up-take from exosomes released in female luminal fluids[69]
2aoviductmus musculusultracentrifugationspermTEMoEVs transfer increase PMCA1 and PMCA4 activity in sperm[70]
2aoviductfelis catuspolymer precipitationSpermNTAoEVs contain protein related to energy metabolism, sperm functionality and enhance sperm motility and fertility in vitro[71]
2aoviductbos taurusultracentrifugationSpermDLSoEVs induced acrosome reaction and signalingevents associated with sperm capacitation[72]
2boviductbos taurusultracentrifugationEmbryoTEMoEVs were internalized in embryo, increasing blastocyst rate and embryo quality[73]
2boviductbos taurusultracentrifugationEmbryoNTA, TEMoEVs increased embryo quality and altered expression of SNRPN[74]
3follicular fluid bos taurusultracentrifugationEmbryotRPS, TEMFF isolated EVs caused transcription and epigenetic alteration in embryos[75]
4embryosus scrofaultracentrifugation and precipitationendometrial epithelial cellsTEMevidence on embryo endometrial cross-talk mediated by EVs. EVs released by trophectoderm-cells increase the expression of miRNAs in maternal endothelial cells related to angiogenesis signaling[76]
4uterine flushings (UFs) from pregnant ewesovis ariespolymer precipitationendometrial epithelial cellsTEM, NTAConceptus-derived EVs induce the expression of Interferon-Stimulated Genes ISG in bovine EECs culture [77]
5cytotrophoblast cell-derived exosomehomo sapiensultracentrifugationextravillous trophoblasts (EVT)TEMExosomes regulate intercellular communication between placental cells and EVT cell invasion in an oxygen-dependent manner[78]
6aEndometrial tissue and uterine fluidhomo sapiensultracentrifugationEmbryotRPS, FCEndometrial derived EVs contain specific miRNA that may contribute to the endometrial-embryo cross talk and embryo implantation[1]
6aendometrial epithelial cellhomo sapiens/mus musculusultracentrifugationEmbryoTEMEndometrial derived EVs transport miRNAs influencing embryo transcriptomic for genes related to embryonic adhesion phenomenon[8]
6aUterine Fluid bos tauruspolymer precipitationEmbryoTEM, NTAEVs from uterine fluid regulate bovine conceptus implantation[79]
6aUterine Fluidbos tauruspolymer precipitationEmbryoEMEVs from uterine fluid of cows with endometritis impact blastocyst development[80]
6bUterine Fluidgallus gallusultracentrifugationSpermTEMUterine fluid EV contain proteins that may play an essential role in the preservation of sperm functions[81]
Table 3. Studies reporting the use of molecular cargos of EVs as biomarkers in reproductive medicine. Studies were categorized by different pathological conditions describing the source and method of EV isolation and the main results obtained.
Table 3. Studies reporting the use of molecular cargos of EVs as biomarkers in reproductive medicine. Studies were categorized by different pathological conditions describing the source and method of EV isolation and the main results obtained.
Biomarkers for:EVs Isolated fromSpeciesIsolation MethodMain ResultsReferences
female reproductive cancerserumhomo sapienspolymer precipitationEV miRNAs increase in the serum of epithelial ovarian cancer patients [123]
female fertilityfollicular fluid homo sapiensultracentrifugationEV-miRNAs in follicular fluid are associated with urinary concentrations of phenols and phthalate metabolites[124]
female fertilitymedium of blastocysts and endometrial cell co-cultureshomo sapienspolymer precipitationEV-bound secreted miRNAs are altered in co-culture experiments with blastocysts and endometrial cells isolated from patients diagnosed with AMA or endometriosis[125]
embryo qualitymedium of embryo cultureshomo sapiensno isolationDNA content in EVs isolated from embryo culture is linked to successful implantation[126]
placenta qualityprimary cytotrophoblasts and serumhomo sapienspolymer precipitation and ultracentrifugation serum EVs from patients with preeclampsia showed alteration in syncytin content[127]
placenta qualityplasmahomo sapiensultracentrifugationmicroRNAs from plasma EVs are altered in preeclampsia[128]
placenta qualityplasmahomo sapienspolymer precipitation and ultracentrifugation EVs from preeclampsia patients delivered antiangiogenic factors to endothelial cells[102]
placenta qualityplasmahomo sapiensUltracentrifugation and size exclusion chromatographyProteomic analysis of plasma EVs revealed protein alterations related to gestational diabetes mellitus[129]
placenta qualitycondition media of chorionic villihomo sapiensUltracentrifugation Gestational diabetes mellitus alters miRNA profile of EVs isolated from chorionic villi explant cultures[130]
early abortionserumbos taurusUltracentrifugation EVs from serum contain microRNAs related to embryonic mortality in cows[131]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Capra, E.; Lange-Consiglio, A. The Biological Function of Extracellular Vesicles during Fertilization, Early Embryo—Maternal Crosstalk and Their Involvement in Reproduction: Review and Overview. Biomolecules 2020, 10, 1510. https://doi.org/10.3390/biom10111510

AMA Style

Capra E, Lange-Consiglio A. The Biological Function of Extracellular Vesicles during Fertilization, Early Embryo—Maternal Crosstalk and Their Involvement in Reproduction: Review and Overview. Biomolecules. 2020; 10(11):1510. https://doi.org/10.3390/biom10111510

Chicago/Turabian Style

Capra, Emanuele, and Anna Lange-Consiglio. 2020. "The Biological Function of Extracellular Vesicles during Fertilization, Early Embryo—Maternal Crosstalk and Their Involvement in Reproduction: Review and Overview" Biomolecules 10, no. 11: 1510. https://doi.org/10.3390/biom10111510

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