Next Article in Journal
Fatty Acid Binding Protein 3 Enhances the Spreading and Toxicity of α-Synuclein in Mouse Brain
Next Article in Special Issue
Asynchronous Embryo Transfer Followed by Comparative Transcriptomic Analysis of Conceptus Membranes and Endometrium Identifies Processes Important to the Establishment of Equine Pregnancy
Previous Article in Journal
Aromatase Inhibitors as Adjuvant Treatment for ER/PgR Positive Stage I Endometrial Carcinoma: A Retrospective Cohort Study
Previous Article in Special Issue
Physiologic Events of Embryo Implantation and Decidualization in Human and Non-Human Primates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MiRNAs in the Peri-Implantation Period: Contribution to Embryo–Maternal Communication in Pigs

by
Monika M. Kaczmarek
1,*,
Joanna Najmula
1,
Maria M. Guzewska
1 and
Emilia Przygrodzka
2
1
Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
2
University of Nebraska Medical Center, Omaha, NE 68198, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(6), 2229; https://doi.org/10.3390/ijms21062229
Submission received: 11 December 2019 / Revised: 29 January 2020 / Accepted: 30 January 2020 / Published: 23 March 2020
(This article belongs to the Special Issue Embryo-Maternal Interactions Underlying Reproduction in Mammals)

Abstract

:
MicroRNAs (miRNAs) constitute a large family of noncoding RNAs, approximately 22 nucleotides long, which function as guide molecules in RNA silencing. Targeting most protein-coding transcripts, miRNAs are involved in nearly all developmental and pathophysiological processes in animals. To date, the regulatory roles of miRNAs in reproduction, such as fertilization, embryo development, implantation, and placenta formation, among others, have been demonstrated in numerous mammalian species, including domestic livestock such as pigs. Over the past years, it appeared that understanding the functions of miRNAs in mammalian reproduction can substantially improve our understanding of the biological challenges of successful reproductive performance. This review describes the current knowledge on miRNAs, specifically in relation to the peri-implantation period when the majority of embryonic mortality occurs in pigs. To present a broader picture of crucial peri-implantation events, we focus on the role of miRNA-processing machinery and miRNA–mRNA infarctions during the maternal recognition of pregnancy, leading to maintenance of the corpus luteum function and further embryo implantation. Furthermore, we summarize the current knowledge on cell-to-cell communication involving extracellular vesicles at the embryo–maternal interface in pigs. Finally, we discuss the potential of circulating miRNAs to serve as indicators of ongoing embryo–maternal crosstalk.

1. Introduction

Early pregnancy in domestic animals, such as pigs, begins with the period of development of the embryo(s) in the oviduct to reach the blastocyst stage in the uterus. After hatching from the zona pellucida, the blastocyst elongates to increase its physical contact area with the endometrium and starts an intensive dialogue with the uterus, establishing the foundation for a successful pregnancy. As in other mammals, conceptus development requires the uterus and is concomitant with a maternal recognition of pregnancy (days 10–13 post-estrus) via embryonic signaling to maintain the corpus luteum (CL) function (i.e., progesterone (P4) production) as well as the onset of implantation (days 14–19) and further placenta formation.
In pigs, two main embryonic signals—estradiol-17beta (E2) and prostaglandin E2 (PGE2)—prevent regression of the CL, which occurs on days 13–14 of the estrous cycle. CL rescue is achieved by a paracrine action of the conceptus on the endometrium and the luteal tissue to stop the beginning of luteolysis [1]. Reciprocal communication between the preimplantation conceptus not only results in maintaining P4 production by the ovaries but also sets up the environment for successful embryonic development and implantation.
Uterine receptivity for conceptus elongation and implantation develops during each cycle in response to P4 and is modified in response to products secreted by the conceptus and endometrium sensed by the competent embryo. Preparation of the uterus synchronized with the development of the embryo is necessary to establish proper communication between the embryo and the mother during early pregnancy. Establishing firm connections between the endometrium and the embryo requires a specific mutual dialogue achieved through the secretion of steroid hormones as well as a number of different agents, such as growth factors, prostanoids, and cytokines [2,3]. Importantly, the growing conceptus has to communicate with the endometrium not only to appose, attach, and implant, but also to sustain favorable conditions for its own growth. Recently, microRNAs (miRNAs) have emerged as new players in the fine-tuning of the endometrial milieu, embryo development, and implantation in mammals via posttranscriptional gene regulation mechanisms.
In this review, we summarize recent progress in miRNA-mediated regulations of the peri-implantation period in pigs and highlight the significance of miRNAs in different aspects of successful pregnancy outcomes. First, we discuss the importance of miRNA-processing machinery during early pregnancy. Second, we focus on the dynamics of peri-implantation events, looking at miRNAs at the embryo–maternal interface and in the CL during pregnancy. Furthermore, we summarize the role of miRNA transport at the embryo–maternal interface involving extracellular vesicles (EVs). Finally, we discuss an intriguing characteristic of miRNAs—their presence in circulation—and provide a set of conclusive remarks to guide future studies.

2. Overview of MiRNA Biogenesis and Function

MiRNAs are a class of endogenous, small non-coding RNAs, with an average length of 22 nucleotides. Slightly over half of all currently identified miRNAs are intergenic, located in noncoding transcription units, while the remaining miRNAs are intragenic, processed mostly from introns and relatively few exons of protein-coding genes [4,5]. In addition, miRNAs can occur in clusters, which means that two or more miRNAs are simultaneously transcribed from adjacent miRNA genes [6]. This intriguing family of regulatory molecules is mostly transcribed by RNA polymerase II from DNA sequences into primary miRNAs (pri-miRNAs), containing a local stem–loop structure that encodes miRNA sequences in the arm of the stem. Pri-miRNAs are processed by a DROSHA/DGCR8 microprocessor complex into 70–100 nt hairpin/stem–loop precursors (pre-miRNAs). Once exported from the nucleus by exportin-5 (XPO5), pre-miRNAs are cleaved by DICER1 (RNase III endonuclease) to produce RNA duplexes approximately 22 nt long, one strand of which is selected and incorporated into the effector complex, RNA-induced silencing complex (RISC) [7,8]. DICER1 and its protein partners, HIV-1 transactivation response (TAR), RNA-binding protein 2 (TARBP2), a trinucleotide repeat containing 6A (TNRC6A), and members of the Argonaute (AGO1–4) family, are the main components of this complex. RISC binds to the target mRNA in a sequence-specific manner and induces mRNA cleavage, translational inhibition, or cleavage-independent mRNA degradation [9,10]. Although the aforementioned canonical pathway accounts for the production of most miRNAs, it has also been shown that alternative (non-canonical) pathways for miRNA biogenesis exist, which bypass part of the abovementioned biogenesis steps [11]. In most cases, miRNAs interact with the 3′ UTR of target mRNAs to suppress expression [12]. However, miRNA interactions with other regions, such as the 5′ UTR or coding sequence, have also been reported [13]. While a single miRNA may target many genes, the expression of a particular gene can be modulated by multiple miRNAs [14]. Interestingly, miRNAs belonging to the same genomic cluster can share targets between specific signaling pathways [6], which consequently can enhance miRNAs’ mediated effects on biological functions.

3. MiRNA Biogenesis at the Embryo-Maternal Interface

Several reports have suggested that the disrupted expression of genes involved in miRNA synthesis or transport, mainly DICER1 and AGO2, may have a serious effect on reproductive functions or embryo development, most likely due to altered miRNA levels [15,16]. The importance of DICER1 expression in early embryos has already been reported in animals, including domestic livestock such as pigs [17] and cattle [18]. Notably, knockout of Dicer1 in mouse causes morphologic abnormalities and stunted growth in embryonic day (E) 7.5 embryos and lethality by E11.5 [19], clearly showing its crucial role in development. Among others, the loss of Dicer in the mouse reproductive tract (Dicer Amhr2-cKO) compromised uterine development, leading to pregnancy loss following the wild-type embryo transfer [20]. Interestingly, simultaneous estrogen and P4 treatment in mice increased the uterine expression of DROSHA, DGCR8, XPO5, and DICER1 [21], suggesting involvement of steroid hormones in miRNA biogenesis.
Recently, pregnancy stage-dependent expression profiles of genes involved in miRNA synthesis and transport were demonstrated in porcine conceptuses, trophoblasts, and endometria [22,23]. Uterine glands and luminal epithelia were the only sites of DICER1 and AGO2 immunoreactivity, where miRNA biogenesis seems likely to occur during early pregnancy in pigs. Remarkably, AGO4 and DGCR8 endometrial expression significantly increased on day 16 of pregnancy, concomitantly with a slight increase of DICER1 and AGO3. Conceptuses, however, showed increased expression of XPO5, DICER1, TNRC6A, and AGO2 on days 11–12, when porcine blastocysts undergo rapid and radical morphological changes from spherical, through tubular, to filamentous forms, accompanied by increasing transcriptomic activity and E2 secretion [24,25]. Characteristic, dynamic gene expression patterns involved in miRNA biogenesis and transport observed in endometria and developing conceptuses on days 10–20 of pregnancy point at the crucial involvement of miRNAs in the regulation of peri-implantation events in pigs.

4. MiRNAs at the Embryo—Maternal Interface

The key phenomena of porcine early pregnancy are the prolonged embryo migration throughout the uterus and the following epitheliochorial placentation when maternal and fetal layers lay in close contact without invading each other [26]. At this stage, a great variety of molecules is exchanged between both parties [3]. Consequently, dramatic transcriptome changes can be observed, and thousands of genes are differentially expressed at this time in both contacting tissues [27,28,29,30]. Those very time-specific transcriptomic changes demand precise regulations that, among others, are driven by miRNAs.
To date, numerous miRNAs have been detected at different stages of porcine embryo development, ranging from zygotes and early embryos [17,31], through peri-implantation embryos and its trophoblasts [22,32,33,34], to placentas on day 90 of gestation ([35,36]; see examples in Table 1).
Comparisons of 8-cell embryos and day 7 blastocysts revealed 13 miRNAs, differentiating both developmental stages. Their potential targets were overrepresented in Gene Ontology categories, such as cell motion and migration, helicase activity, chromatin organization, transcriptional activity, and cellular component enrichment of Golgi cis cisternae—processes that are crucial during the formation of blastocysts, when cells rapidly proliferate, differentiate, and migrate to create the inner cell mass and trophectoderm [17]. Later stages of porcine embryo development were studied using high-throughput sequencing or microarrays and revealed hundreds (368 and 220, respectively) of miRNAs expressed during the peri-implantation period of pregnancy [22,34]. Both studies found that the identified miRNAs are potential regulators of important processes for embryo development and maternal-embryo interaction (e.g., TGF-beta and p53 signaling, cellular motility, and gonadotropin-releasing hormone signaling).
The miRNAome of the endometrium during pregnancy was widely characterized by microarrays [33,37] and sequencing [23]. MiRNAs differentially expressed between days 15 and 50 of gestation were mostly involved in endometrial remodeling (e.g., cell proliferation, migration, apoptosis, cytoskeleton organization, and blood vessel development) and cell communication (e.g., response to hormone stimulus, and cell-to-cell and cell-to-matrix adhesion) [37]. Using different methods, the set of miRNAs worth noticing is consistently shown by two groups, as differentially expressed between day 20 pregnant vs. cyclic endometrium: miR-27a, miR-30d, miR-205, and miR-574 [23,33].
Intriguingly, porcine endometria [23] as well as blastocysts [38] are abundant not only in canonical miRNAs but also isomiRs—widely reported variants of miRNAs—which have been proven to work cooperatively [43]. Many miRNAs can be co-expressed with their isomiRs and/or other members of the family (e.g., 23a/b, 148a/b in the porcine endometrium), resulting in wider or stronger biological effects. Although the role of a vast reservoir of isomiRs in reproductive tissues remains unclear, it was proven that transfection with either miR-140-3p, its isomiR, or both simultaneously added significantly downregulated putative target gene expression (i.e., ACVR2B, KCNMA1, SIRT1, LIF) in porcine stromal cells [23].
The key molecules of maternal recognition of pregnancy in pigs are estrogens (mainly E2) secreted by conceptuses during rapid elongation [44]. For years, it has been known that disrupted estrogen exposure can lead to pregnancy abnormalities [45,46]. Interestingly, uterine miRNA biogenesis components were shown to be under steroidal regulation in rodents [21]. Our studies suggested a similar regulatory relationship in the porcine endometrium [23]. However, neither endometrium [47] nor blastocyst [38] miRNAome was shown to be affected on day 10 of pregnancy by E2 administered daily to sows in different doses, even though its concentrations significantly increased in the endometrium.
Differences in endometrial and/or conceptus miRNA expression can be strain-specific and related to the arrest of embryo development and litter size. The peri-implantation endometria of Yorkshire and Chinese Meishan breeds that vary in litter size were reported to differ in miRNA expression. MiRNAs differentially expressed between both stains around day 12 of pregnancy were involved in the regulation of p53 and Wnt signaling pathways, which are both crucial for proper implantation in pigs [48]. When miRNA expressions of trophoblasts of healthy and arrested embryos were compared, only 3 (miR-15b-5p, miR-18a, and miR-222) out of 11 detected miRNAs were differentially expressed, similar to the matching endometrial biopsies (miR-20b, miR-17-5p, and miR-18a) [32]. Indeed, miR-18a has already been associated with human fetal macrosomia [49] and preeclampsia [50]. Recently, it was shown that a single-nucleotide polymorphism (SNP) within a miR-18a binding site within CDC42 3′UTR lowers the ability of this miRNA to regulate and influence litter size by affecting oocyte maturation and placental development in pigs [51]. In addition, an SNP within a miR-27a gene was also found to be associated with pregnancy outcome. Specifically, a T/C mutation in a coding sequence of miR-27a was associated with litter size, and, as suggested by the authors, could serve as a molecular marker for litter size in pig breeding programs [52].
Early pregnancy events are accompanied by intensive changes in the mother’s immune system. Top-ranked processes affected by differentially expressed miRNAs in trophoblasts between days 12 to 20 of pregnancy indicate their role in inflammatory processes and cell-mediated immune response. The number of inflammatory mediators can be potentially targeted by miRNAs detected in endometria [23] and trophoblasts [22]. Eventually, downregulation of LIF and its receptor in endometrial luminal epithelial cells by miR-125b was validated in vitro [22]. In the endometrium following insemination and during different stages of early pregnancy, there are noticeable variations in the distribution of immune cells [53]. Analysis of miRNAs in the lymphocytes of endometria in days 20 and 50 of gestation revealed a set of miRNAs involved in immune cell development at the fetal–maternal interface. Interestingly, miR-150, miR-296-5p, and miR-19a were differentially expressed in endometria with arrested and healthy embryos on day 20. Since most of these miRNAs are involved in angiogenesis and immune cell development, the authors suggested that arrested conceptuses try to compensate for their potential lack of angiogenesis and/or other processes essential for survival [32].

5. MiRNAs Carried by EVs at the Embryo–Maternal Interface

For the first time, small vesicles along the interface between the porcine endometrium and developing conceptuses were detected on day 16 of pregnancy by electron microscopy in the early 1990s of the last century [54]. This observation was the first hint that EVs can be active mediators of embryo–maternal communication during early pregnancy in pigs. To date, EVs have been isolated from different body fluids in different species, including porcine seminal plasma [55] and uterine lumen flushings [22]. Recently, intensive secretion of EVs by endometrial and trophoblast cells was detected during the peri-implantation period in pigs. A large population of EVs of different sizes was observed in the lumen of uterine glands, along with a number of multivesicular bodies located in the cytoplasm of luminal and glandular epithelia as well as trophoblast cells [56]. This is consistent with the current knowledge that EVs can be secreted by most cells in physiological and pathological conditions, in response to different stimuli and changes in the extracellular environment [57].
EVs are a heterogeneous population of biological particles that are surrounded by a double phospholipid membrane. EVs have been divided into two groups, according to their size: microvesicles/ectosomes (100–1000 nm), budding directly out from the plasma membrane; and exosomes (30–100 nm), intraluminal vesicles created during maturation of multivesicular bodies released from the originating cell via exocytosis [58]. The endosomal sorting complex required for transport (ESCRT) catalyzes the formation of multivesicular bodies [59]. The ESCRT complex is composed of approximately 30 proteins that assemble into four complexes, ESCRT-0, -I, -II, and -III. Subunits of ESCRT recognize ubiquitylated cargo, deform endosomal membranes, and catalyze intraluminal vesicle formation with sorted cargo [60]. EVs have the ability to carry different molecules such as proteins, DNA, various RNA forms, including miRNAs, and other cellular and membrane components (e.g., tetraspanins CD63, CD9), contributing to intercellular communication [61]. Trafficking routes in the generation of EVs also involve RAS-related protein (RAB) GTPases, master regulators of intracellular membrane traffic [58]. To date, the organization and activity of the ESCRT complex and vesicle trafficking have not yet been studied during early pregnancy in pigs. Our preliminary results suggest that reproductive status (pregnant, non-pregnant) affects the expression of ESCRT and RAB family members in the endometrium and trophoblast during peri-implantation in pigs [62].
The main functions of EVs depend on their ability to interact with recipient cells and to deliver their content. Each cell type may use different pathways of EV uptake, but generally, uptake seems to be a complex and cooperative process involving a combination of different endocytosis pathways. EVs can use receptors to enter target cells and stimulate them via a ligand–receptor interaction, guided by adhesion molecules such as integrins or tetraspanins [58]. Clathrin-dependent endocytosis controls the internalization of many receptors and signal transduction proteins implicated in many key cellular functions such as growth, differentiation, migration, or proliferation [63]. Morphologically, the first stage of clathrin-dependent endocytosis starts through the membrane remodeling process. The membrane deforms into clathrin-coated pits, and invaginated vesicles detach after cargo selection and clathrin uncoating to move along microtubules in the cytoplasm. We observed this mechanism using transmission electron microscopy during the peri-implantation period in pigs. On day 16 of pregnancy, at the apical site of trophoblast cells, we confirmed the uptake of EVs via clathrin-maintained endocytosis (Figure 1). Notably, this pathway of EVs uptake has not yet been analyzed in relation to the cell-to-cell commutation at the embryo–maternal interface during early pregnancy in pigs and other species.
As described before, EVs can horizontally transfer mRNA and miRNA to target cells, which then trigger the translation of functional proteins or induce gene silencing [64]. The involvement of endometrial EVs carrying unique miRNAs in embryo-maternal crosstalk at implantation has already been suggested for several species, including pigs [22,39], sheep [65], and humans [66]. The first discovery of EVs that carry miRNAs in the uterine environment of pigs on days 14 and 16 of pregnancy reinforced the idea of the horizontal transfer of miRNAs between the endometrium and conceptuses in order to support growth and development, implantation, and placenta formation. Indeed, miR-125b identified in uterine lumen-derived EVs delivered to day 12 endometrial luminal epithelial cells, the first site of signal exchange between the mother and conceptus, affected the expression of genes important during pregnancy, such as LIF and LIFR [22]. In addition, EVs secreted in vitro by porcine trophectoderm cells were shown to carry miRNAs (e.g., miR-16, miR-17-5p, miR-20a) and stimulate proliferation of porcine aortic endothelial cells, indicating the possibility of EVs–miRNA mediated effects on the regulation of placental angiogenesis [39]. All of the abovementioned data clearly showed that EVs are active mediators of two-way communication between the embryo and the mother during the peri-implantation period.

6. MiRNAs in Pregnant Corpus Luteum

The fundamental role of miRNAs in ovarian function was proved by knockout of DICER, which inhibited follicle growth, and reduced the ovulation rate and faulty oocyte development [16,67,68]. Further studies showed a number of differentially expressed miRNAs in the bovine and ovine CL during development, regression, or its rescue during pregnancy [69,70,71,72,73,74]. Interestingly, a greater amount of miRNAs was found in the mature than in the developing CL, indicating their potential role in the maintenance of luteal function. Nevertheless, not many studies have provided information on the specific roles of selected miRNA–mRNA interactions on the function of luteal cells, while only a few have shown changes in the expression of miRNAs occurring in the CL during early pregnancy [40,72].
Recently, 14 differentially expressed miRNAs were identified in porcine CL collected right after maternal recognition of pregnancy (day 14 of pregnancy) and luteolysis (day 14 of the estrous cycle) [40]. Overall, seven miRNAs were upregulated (e.g., miR-21a-3p, miR-345-3p, miR-371-5p), and seven miRNAs were downregulated (e.g., miR-181a, miR-532-3p, miR-99b) in the porcine CL on day 14 of the estrous cycle compared to the corresponding day of pregnancy. Interestingly, among miRNA targets upregulated during pregnancy, genes encoding known regulators of luteolysis were found (e.g., EDN1, FOS, JUN, PTGS2, ESR2) [75,76,77,78], while miRNAs elevated in the CL during luteolysis could target genes associated with luteal function maintenance (e.g., PGR, VEGFR1, CREB, PTGER2). Further in silico analysis suggested that miRNAs highly expressed in the CL during early pregnancy can be involved in the cell cycle, cell death and survival, cellular development, growth, and proliferation—events characterizing the time when a decision is made either to regress or maintain the luteal function. Notably, the expression of miRNAs was examined in the whole luteal tissue. Therefore, the aforementioned biological processes can be potentially regulated by differentially expressed miRNAs in various cells forming the porcine CL, such as the endothelial cells, immune cells, or fibroblasts, and not only the luteal cells. Other studies performed on the bovine CL identified 15 miRNAs differentially expressed on day 18 of pregnancy vs. the corresponding day of the estrous cycle, while among predicted targets of these miRNAs, genes involved in immune-related events and apoptosis were found [72].
Previously, the high expression of two clusters, miR-183-96-182 and miR-212-132, was noticed in the luteal vs. follicular tissue of cows, while miR-96 was found to be an important regulator of steroidogenesis and cell survival [74]. In pigs, miRNAs upregulated in the CL of pregnant animals belong to three independent clusters: miR-99b, miR-532, and miR-181a [40]. Again, among targets of miRNAs occurring in the same cluster (i.e., miR-532 and miR-99b), crucial regulators of luteolysis were found, including, for example, EDN1, FOS, JUN, NR4A1, OXTR, and PTGS2 [75,76,78]. Interestingly, identified clusters are conserved or broadly conserved among different animal species and humans; thus, it is possible that some of them can support luteal function in other species as well, especially since miRNAs belonging to cluster miR-99b were previously detected among mostly abundant miRNAs in the mature bovine CL [73].
Luteotrophic and/or luteolytic factors were suggested to be effective modulators of miRNA expression [74,79]. In pigs, incubation of luteal tissue slices with E2 stimulated the expression of miRNAs belonging to the miR-99b cluster [40]. Further, bioinformatics analysis showed multiple SP1 and ESR1 binding sites in the promoter of the miR-99b cluster. SP1 is a zinc finger transcription factor that acts synergistically with ESR1, one of the genomic receptors of estradiol, in the regulation of gene transcription. Therefore, we suggest that E2 increases the expression of miR-99b by the ESR1-SP1 pathway in the CL during early pregnancy. Remarkably, the aforementioned effects of E2 on the expression of the miRNA-99b cluster were concomitant with the inhibitory effect of E2 on the expression of NR4A1 and AKR1C1 in luteal tissue slices [40]. NR4A1 is encoded by immediate-early response genes, which can induce apoptosis or elevate transcription of AKR1C1, an enzyme responsible for the metabolism of progesterone [80,81]. NR4A1 was identified as a marker of luteolysis in the CL of cows and rats [80,81,82]. Our further studies showed that the transfection of luteal tissue slices with mimics of miRNAs belonging to the miR-99b cluster decreases the expression of NR4A1 and AKR1C1, and increases the production of progesterone by luteal tissue slices of pigs [40]. Therefore, it seems likely that E2 can induce the expression of the miR-99b cluster in porcine CL during the peri-implantation period, leading to decreased expression of genes involved in luteolysis (Figure 2).

7. Circulating MiRNAs during Early Pregnancy

MiRNAs have been detected in several extracellular fluids, including plasma and serum [83,84], showing great stability due to their resistance to endogenous ribonucleases [85]. Their stability in blood might also be related to the fact that they are usually chaperoned by various carriers such as RNA-binding proteins like AGO2 [86] or lipoprotein complexes [87], or are packed in EVs [88]. To date, miRNAs have been shown to change significantly throughout the course of many diseases and physiological conditions, including pregnancy disorders [89]. Therefore, they have emerged as potent biomarkers for the diagnosis and prognosis of a disease in a minimally invasive way.
The characteristic composition of circulating miRNAs has been shown in several animals, including domestic animals, during normal and complicated pregnancies. For example, miR-26a levels were higher in 3-week pregnant heifers compared with sham-inseminated counterparts [90]. Broadly conserved miRNAs (e.g., let-7c, miR-143) were found to be signatures of pregnancy in cows; however, elevated levels of these miRNAs were observed during late pregnancy [91,92]. Interestingly, circulating EV-derived miRNAs (e.g., miR-125b-2, let-7c) were associated with increased embryonic mortality on day 17 of pregnancy in cows [93]. However, EV-derived and broadly conserved miR-195 was maintained at a high level in pregnant mares on day 11 [94].
Recently, we detected three pregnancy-specific circulating miRNAs in maternal circulation immediately after initiation of implantation [41]. Increased levels of circulating miRNAs, previously reported to be expressed in either conceptuses (miR-26a and miR-125b [22]) or pregnant endometria (miR-23b [23]), were detected on day 16 of pregnant pigs. Common biological processes and pathways affected by the three miRNAs (e.g., Wnt/beta-catenin, TGF-beta, and FGF signaling), as well as the number of targets (e.g., IFNG, LIFR, FGFR2, TGFBR1, ESR1, SP1) with important roles during pregnancy, were identified in silico, supporting the concept that we may indeed observe signs of ongoing embryo–maternal crosstalk in the circulation of the pregnant pig. Further results showed a plethora of reproductive status-dependent miRNAs, allowing to distinguish between pregnant and non-pregnant animals on days 16 and 20 of pregnancy [42]. Interestingly, serum samples were enriched with miRNAs involved in processes occurring during early pregnancy, such as angiogenesis, embryonic cell proliferation, and differentiation. In addition, analyzed serum samples contained both EVs and AGO2 proteins, both involved in miRNA transportation and cell-to-cell communication. Our results support the possibility of using circulating miRNAs that differ in abundance between cyclic and pregnant pigs as potential indicators of reproductive status during breeding management.

8. Concluding Remarks and Future Directions

Clearly, miRNAs can be now recognized as powerful gene expression regulators during the peri-implantation period in livestock, such as pigs. We and others have clearly shown that miRNAs are present at the embryo–maternal interface and can target genes supporting early pregnancy events, such as embryo development and implantation (Figure 3). However, recent experiments have mostly focused on transcriptomic screening and in silico miRNA–mRNA interaction predictions, and only a few have shown experimentally the impact of particular miRNAs on the trophoblast or endometrial function. The latter observations revealed, however, the complexity of miRNA-mediated gene regulation during early pregnancy in pigs. Still, our understanding of when and how miRNAs can exert regulatory effects on transcription of the embryo–maternal interface is very limited. Deeper investigation is needed to understand which factors contribute to the activity of miRNAs in trophoblasts and endometria, including cell-specific and subcellular location, miRNA and target abundance, miRNA–mRNA binding affinity, miRNA cluster cooperation, and many more related to the cellular context-dependent response.
Recent discoveries of EVs released by both the endometrium and embryos revealed an additional layer to cell-to-cell communication during the peri-implantation period. The conducted research gives an initial picture of the possible embryo–maternal interactions involving EVs. However, many aspects have not yet been investigated, including biogenesis of EVs and packing of the specific molecular cargo in response to different stimuli. Thus far, no mechanism has been described to guide the EVs to deliver selected cargo to target cells at the embryo–maternal interface.
Overall, the latest research definitely extends our knowledge about the expression and role of miRNAs in the luteal tissue. However, more efforts should be made to characterize miRNAs that can be unique/common among different animal species or specific for CL cells, the role of specific miRNA–mRNA interactions, and the regulation of miRNA expression by luteotrophic or luteolytic factors at the crucial and decisive moment when live embryos are present in the uterus.
Cortez and coworkers [95] hypothesized that miRNAs can function as the “oldest” hormones secreted into blood plasma, and in this way, they can exert their effects onto distant parts of the body. Although it seems likely that miRNAs overexpressed during the peri-implantation period are released into the bloodstream from reproductive tissues and developing conceptuses, direct evidence for this or its mechanism is still missing.
Pregnancy monitoring using miRNAs is of particular interest due to often-occurring difficulties in diagnosing complications in the early stages combined with their potentially catastrophic consequences. Research related to detecting and understanding the role of particular miRNAs during the peri-implantation period is noteworthy for animals with high early embryonic mortality, such as pigs. In any species, earlier diagnosis allows for earlier intervention, leading to favorable outcomes.

Author Contributions

J.N., M.M.G., E.P. and M.M.K. wrote the review and approved the final version. Figures were conceived by M.M.G., E.P. and M.M.K. All authors have read and agreed to the published version of the manuscript.

Funding

Some research described in the review was supported by grants funded by the National Science Centre in Poland (2014/15/B/NZ9/04932) and KNOW (Leading National Research Centre) Scientific Consortium “Healthy Animal-Safe Food” (decision of Ministry of Science and Higher Education No. 05-1/KNOW2/2015; grants: KNOW2016/IRZiBŻ/PRO1/01/5, KNOW/2016/IRZiBZ/ESR3/01/4).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

3′ UTRThree prime untranslated region
5′ UTRFive prime untranslated region;
ACVR2BActivin receptor type-2B
AGO1-4Argonaute RISC components 1-4
AKR1C1Aldo-keto reductase family 1 member C1
CDC42Cell division cycle 42
CLCorpus luteum
CREBCyclic AMP-responsive element-binding protein
DGCR8DiGeorge syndrome critical region gene 8
DICER1Dicer 1, ribonuclease III
DROSHADrosha, ribonuclease III
E2Estradiol-17beta
EDN1Endothelin 1
ESCRTEndosomal sorting complex required for transport
ESR1Estrogen receptor 1
ESR2Estrogen receptor 2
EVsExtracellular vesicles
FGFFibroblast growth factor
FGFR2Fibroblast growth factor receptor 2
FOSFos proto-oncogene
IFNGInterferon gamma
JUNJun proto-oncogene
KCNMA1Potassium calcium-activated channel subfamily M Alpha 1
LIFLeukemia inhibitory factor
LIFRLeukemia inhibitory factor receptor
miRNAMicroRNA
NR4A1Nuclear receptor subfamily 4 group A member 1
OXTROxytocin receptor
P4Progesterone
PGE2Prostaglandin E2
PGRProgesterone receptor
PTGER2Prostaglandin E receptor 2
PTGS2Prostaglandin-endoperoxide synthase
RABRAS-related protein GTPases
RISCRNA-Induced Silencing Complex
SIRT1Sirtuin 1
SNPSingle nucleotide polymorphism
TARBP 2HIV-1 transactivation response (TAR), RNA- binding protein 2
TGF-betaTransforming growth factor beta
TGFBR1Transforming growth factor beta receptor 1
TNRC6ATrinucleotide repeat containing 6A
VEGFR1Vascular endothelial growth factor A receptor 1
XPO5Exportin-5

References

  1. Ziecik, A.J.; Przygrodzka, E.; Jalali, B.M.; Kaczmarek, M.M. Regulation of the porcine corpus luteum during pregnancy. Reproduction 2018, 156, R57–R67. [Google Scholar] [CrossRef] [PubMed]
  2. Blitek, A.; Kaczmarek, M.M.; Waclawik, A.; Ziecik, A.J. Embryo-maternal relationships during the peri-implantation period- new and old players. In Control of Pig Reproduction IX.; Rodriguez-Martinez, H., Soede, N.M., Flowers, W.L., Eds.; Nottingham University Press: Nottingham, UK, 2013; pp. 61–70. [Google Scholar]
  3. Waclawik, A.; Kaczmarek, M.M.; Blitek, A.; Kaczynski, P.; Ziecik, A.J. Embryo-maternal dialogue during pregnancy establishment and implantation in the pig. Mol. Reprod. Dev. 2017, 84, 842–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kim, Y.K.; Kim, V.N. Processing of intronic microRNAs. EMBO J. 2007, 26, 775–783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. De Rie, D.; Abugessaisa, I.; Alam, T.; Arner, E.; Arner, P.; Ashoor, H.; Åström, G.; Babina, M.; Bertin, N.; Burroughs, A.M.; et al. An integrated expression atlas of miRNAs and their promoters in human and mouse. Nat. Biotechnol. 2017, 35, 872–878. [Google Scholar] [CrossRef]
  6. Lai, X.; Vera, J. MicroRNA Clusters. In Encyclopedia of Systems Biology; Dubitzky, W., Wolkenhauer, O., Cho, K.H., Yokota, H., Eds.; Springer: New York, NY, USA, 2013; pp. 77–141. [Google Scholar]
  7. Khvorova, A.; Reynolds, A.; Jayasena, S.D. Functional siRNAs and miRNAs exhibit strand bias. Cell 2003, 115, 209–216. [Google Scholar] [CrossRef] [Green Version]
  8. Schwarz, D.S.; Hutvagner, G.; Du, T.; Xu, Z.; Aronin, N.; Zamore, P.D. Asymmetry in the assembly of the RNAi enzyme complex. Cell 2003, 115, 199–208. [Google Scholar] [CrossRef] [Green Version]
  9. Pillai, R.S. MicroRNA function: Multiple mechanisms for a tiny RNA? RNA 2005, 11, 1753–1761. [Google Scholar] [CrossRef] [Green Version]
  10. Valencia-Sanchez, M.A.; Liu, J.; Hannon, G.J.; Parker, R. Control of translation and mRNA degradation by miRNAs and siRNAs. Genes. Dev. 2006, 20, 515–524. [Google Scholar] [CrossRef] [Green Version]
  11. Kim, Y.K.; Kim, B.; Kim, V.N. Re-evaluation of the roles of DROSHA, Export in 5, and DICER in microRNA biogenesis. Proc. Natl. Acad. Sci. USA 2016, 113, E1881–E1889. [Google Scholar] [CrossRef] [Green Version]
  12. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell. Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
  13. Broughton, J.P.; Lovci, M.T.; Huang, J.L.; Yeo, G.W.; Pasquinelli, A.E. Pairing beyond the seed supports microRNA targeting specificity. Mol. Cell. 2016, 64, 320–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sood, P.; Krek, A.; Zavolan, M.; Macino, G.; Rajewsky, N. Cell-type-specific signatures of microRNAs on target mRNA expression. Proc. Natl. Acad. Sci. USA 2006, 103, 2746–2751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Morita, S.; Horii, T.; Kimura, M.; Goto, Y.; Ochiya, T.; Hatada, I. One Argonaute family member, AGO2 (Ago2), is essential for development and appears not to be involved in DNA methylation. Genomics 2007, 89, 687–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hong, X.; Luense, L.J.; McGinnis, L.K.; Nothnick, W.B.; Christenson, L.K. Dicer1 is essential for female fertility and normal development of the female reproductive system. Endocrinol. 2008, 149, 6207–6712. [Google Scholar] [CrossRef] [Green Version]
  17. Stowe, H.M.; Curry, E.; Calcatera, S.M.; Krisher, R.L.; Paczkowski, M.; Pratt, S.L. Cloning and expression of porcine Dicer and the impact of developmental stage and culture conditions on MicroRNA expression in porcine embryos. Gene 2012, 501, 198–205. [Google Scholar] [CrossRef]
  18. Burrola-Barraza, M.E.; Hernández-Seáñez, R.; Barceló-Fimbres, M.; Rodríguez-Almeida, F.A.; González-Rodríguez, E.; García-Quiñónez, S.; Grado-Ahuir, J.A.; Moreno-Brito, V. Dicer gene expression during early bovine embryo development. Mol. Reprod. Dev. 2011, 78, 622. [Google Scholar] [CrossRef]
  19. Bernstein, E.; Kim, S.Y.; Carmell, M.A.; Murchison, E.P.; Alcorn, H.; Li, M.Z.; Mills, A.A.; Elledge, S.J.; Anderson, K.V.; Hannon, G.J. Dicer is essential for mouse development. Nat. Genet. 2003, 35, 215–217. [Google Scholar] [CrossRef]
  20. Gonzalez, G.; Behringer, R.R. Dicer is required for female reproductive tract development and fertility in the mouse. Mol. Reprod. Dev. 2009, 76, 678–688. [Google Scholar] [CrossRef] [Green Version]
  21. Nothnick, W.B.; Healy, C.; Hong, X. Steroidal regulation of uterine miRNAs is associated with modulation of the miRNA biogenesis components Exportin-5 and Dicer1. Endocrine 2010, 37, 265–273. [Google Scholar] [CrossRef] [Green Version]
  22. Krawczynski, K.; Najmula, J.; Bauersachs, S.; Kaczmarek, M.M. MicroRNAome of porcine conceptuses and trophoblasts: Expression profile of microRNAs and their potential to regulate genes crucial for establishment of pregnancy. Biol. Reprod. 2015, 92, 21. [Google Scholar] [CrossRef]
  23. Krawczynski, K.; Bauersachs, S.; Reliszko, Z.P.; Graf, A.; Kaczmarek, M.M. Expression of microRNAs and isomiRs in the porcine endometrium: Implications for gene regulation at the maternal-conceptus interface. BMC Genom. 2015, 16, 906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Geisert, R.D.; Renegar, R.H.; Thatcher, W.W.; Roberts, R.M.; Bazer, F.W. Establishment of pregnancy in the pig: I. Interrelationships between preimplantation development of the pig blastocyst and uterine endometrial secretions. Biol. Reprod. 1982, 27, 925–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Geisert, R.D.; Brookbank, J.W.; Roberts, R.M.; Bazer, F.W. Establishment of pregnancy in the pig: II. Cellular remodeling of the porcine blastocyst during elongation on day 12 of pregnancy. Biol. Reprod. 1982, 27, 941–955. [Google Scholar] [CrossRef] [PubMed]
  26. Dantzer, V. Electron microscopy of the initial stages of placentation in the pig. Anat. Embryol. 1985, 172, 281–293. [Google Scholar] [CrossRef]
  27. Ross, J.W.; Ashworth, M.D.; Stein, D.R.; Couture, O.P.; Tuggle, C.K.; Geisert, R.D. Identification of differential gene expression during porcine conceptus rapid trophoblastic elongation and attachment to uterine luminal epithelium. Physiol. Genom. 2009, 36, 140–148. [Google Scholar] [CrossRef] [Green Version]
  28. Østrup, E.; Bauersachs, S.; Blum, H.; Wolf, E.; Hyttel, P. Differential endometrial gene expression in pregnant and nonpregnant sows. Biol. Reprod. 2010, 83, 277–285. [Google Scholar] [CrossRef] [Green Version]
  29. Franczak, A.; Wojciechowicz, B.; Kotwica, G. Transcriptomic analysis of the porcine endometrium during early pregnancy and the estrous cycle. Reprod. Biol. 2013, 13, 229–237. [Google Scholar] [CrossRef]
  30. Kiewisz, J.; Krawczynski, K.; Lisowski, P.; Blitek, A.; Zwierzchowski, L.; Ziecik, A.J.; Kaczmarek, M.M. Global gene expression profiling of porcine endometria on Days 12 and 16 of the estrous cycle and pregnancy. Theriogenology 2014, 82, 897–909. [Google Scholar] [CrossRef]
  31. Yang, C.-X.; Du, Z.-Q.; Wright, E.C.; Rothschild, M.F.; Prather, R.S.; Ross, J.W. Small RNA profile of the cumulus-oocyte complex and early embryos in the pig. Biol. Reprod. 2012, 87, 117. [Google Scholar] [CrossRef]
  32. Bidarimath, M.; Edwards, A.K.; Wessels, J.M.; Khalaj, K.; Kridli, R.T.; Tayade, C. Distinct microRNA expression in endometrial lymphocytes, endometrium, and trophoblast during spontaneous porcine fetal loss. J. Reprod. Immunol. 2015, 107, 64–79. [Google Scholar] [CrossRef]
  33. Wessels, J.M.; Edwards, A.K.; Khalaj, K.; Kridli, R.T.; Bidarimath, M.; Tayade, C. The microRNAome of pregnancy: Deciphering miRNA networks at the maternal-fetal interface. PLoS ONE 2013, 8, e72264. [Google Scholar] [CrossRef] [PubMed]
  34. Wright, E.C. MicroRNA expression and function during porcine oocyte maturation and early embryonic development. PhD Thesis, Iowa State University, Ames, IA, USA, 2012. Available online: http://lib.dr.iastate.edu/cgi/viewcontent.cgi?article=3529&context=etd (accessed on 31 October 2019).
  35. Su, L.; Zhao, S.; Zhu, M.; Yu, M. Differential expression of microRNAs in porcine placentas on Days 30 and 90 of gestation. Reprod. Fertil. Dev. 2010, 22, 1175–1182. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, R.; Wang, M.; Su, L.; Li, X.; Zhao, S.; Yu, M. The expression pattern of microRNAs and the associated pathways Involved in the development of porcine placental folds that contribute to the expansion of the exchange surface area. Biol. Reprod. 2015, 93, 62. [Google Scholar] [CrossRef] [PubMed]
  37. Su, L.; Liu, R.; Cheng, W.; Zhu, M.; Li, X.; Zhao, S.; Yu, M. Expression patterns of microRNAs in porcine endometrium and their potential roles in embryo implantation and placentation. PLoS ONE 2014, 9, e87867. [Google Scholar] [CrossRef]
  38. Bick, J.T.; Flöter, V.L.; Robinson, M.D.; Bauersachs, S.; Ulbrich, S.E. Small RNA-seq analysis of single porcine blastocysts revealed that maternal estradiol-17beta exposure does not affect miRNA isoform (isomiR) expression. BMC Genomics 2018, 19, 590. [Google Scholar] [CrossRef] [Green Version]
  39. Bidarimath, M.; Khalaj, K.; Kridli, R.T.; Kan, F.W.; 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]
  40. Przygrodzka, E.; Sokołowska, G.; Myszczynski, K.; Krawczynski, K.; Kaczmarek, M.M. Clustered microRNAs: The molecular mechanism supporting the maintenance of luteal function during early pregnancy. FASEB J. 2020. [Google Scholar] [CrossRef] [Green Version]
  41. Reliszko, Z.P.; Gajewski, Z.; Kaczmarek, M.M. Signs of embryo-maternal communication: miRNAs in the maternal serum of pregnant pigs. Reproduction 2017, 154, 217–228. [Google Scholar] [CrossRef] [Green Version]
  42. Reliszko, Z.P.; Myszczynski, K.; Kaczmarek, M.M. Differential expression of circulating miRNAs in maternal serum during early pregnancy in the pig. In Proceedings of the 10th International Conference on Pig Reproduction, Columbia, MO, USA, 11–14 June 2017. [Google Scholar]
  43. Cloonan, N.; Wani, S.; Xu, Q.; Gu, J.; Lea, K.; Heater, S.; Barbacioru, C.; Steptoe, A.L.; Martin, H.C.; Nourbakhsh, E.; et al. MicroRNAs and their isomiRs function cooperatively to target common biological pathways. Genome Biol. 2011, 12, R126. [Google Scholar] [CrossRef] [Green Version]
  44. Geisert, R.D.; Thatcher, W.W.; Roberts, R.M.; Bazer, F.W. Establishment of Pregnancy in the Pig: III. Endometrial secretory response to estradiol valerate administered on Day 11 of the estrous cycle. Biol. Reprod. 1982, 27, 957–965. [Google Scholar] [CrossRef] [Green Version]
  45. Geisert, R.D.; Morgan, G.L.; Zavy, M.T.; Blair, R.M.; Gries, L.K.; Cox, A.; Yellin, T. Effect of asynchronous transfer and oestrogen administration on survival and development of porcine embryos. J. Reprod. Fertil. 1991, 93, 475–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Pope, W.F.; Lawyer, M.S.; Butler, W.R.; Foote, R.H.; First, N.L. Dose-response shift in the ability of gilts to remain pregnant following exogenous estradiol-17 beta exposure. J. Anim. Sci. 1986, 63, 1208–1210. [Google Scholar] [CrossRef] [PubMed]
  47. Flöter, V.L.; Lorenz, A.K.; Kirchner, B.; Pfaffl, M.W.; Bauersachs, S.; Ulbrich, S.E. Impact of preimplantational oral low-dose estradiol-17β exposure on the endometrium: The role of miRNA. Mol. Reprod. Dev. 2018, 85, 417–426. [Google Scholar] [CrossRef] [Green Version]
  48. Li, W.; Xi, Y.; Xue, S.; Wang, Y.; Wu, L.; Liu, H.; Lei, M. Sequence analysis of microRNAs during pre-implantation between Meishan and Yorkshire pigs. Gene 2018, 646, 20–27. [Google Scholar] [CrossRef] [PubMed]
  49. Ge, Q.; Zhu, Y.; Li, H.; Tian, F.; Xie, X.; Bai, Y. Differential expression of circulating miRNAs in maternal plasma in pregnancies with fetal macrosomia. Int. J. Mol. Med. 2015, 35, 81–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Zhu, X.M.; Han, T.; Sargent, I.L.; Yin, G.W.; Yao, Y.Q. Differential expression profile of microRNAs in human placentas from preeclamptic pregnancies vs normal pregnancies. Am. J. Obstet. Gynecol. 2009, 200, 661.e1–661.e7. [Google Scholar] [CrossRef] [PubMed]
  51. Liu, R.; Deng, D.; Liu, X.; Xiao, Y.; Huang, J.; Wang, F.; Li, X.; Yu, M. A miR-18a binding-site polymorphism in CDC42 3′UTR affects CDC42 mRNA expression in placentas and is associated with litter size in pigs. Mamm. Genome 2019, 30, 34–41. [Google Scholar] [CrossRef]
  52. Lei, B.; Gao, S.; Luo, L.F.; Xia, X.Y.; Jiang, S.W.; Deng, C.Y.; Xiong, Y.Z.; Li, F.E. A SNP in the miR-27a gene is associated with litter size in pigs. Mol. Biol. Rep. 2011, 38, 3725–3729. [Google Scholar] [CrossRef]
  53. Bischof, R.J.; Brandon, M.R.; Lee, C.S. Cellular immune responses in the pig uterus during pregnancy. J. Reprod. Immunol. 1995, 29, 161–178. [Google Scholar] [CrossRef]
  54. Keys, J.L.; King, G.J. Microscopic examination of porcine conceptus-maternal interface between days 10 and 19 of pregnancy. Am. J. Anat. 1990, 188, 221–238. [Google Scholar] [CrossRef]
  55. Barranco, I.; Padilla, L.; Parrilla, I.; Álvarez-Barrientos, A.; Pérez-Patiño, C.; Peña, F.J.; Martínez, E.A.; Rodriguez-Martínez, H.; Roca, J. Extracellular vesicles isolated from porcine seminal plasma exhibit different tetraspanin expression profiles. Sci. Rep. 2019, 9, 11584. [Google Scholar] [CrossRef] [Green Version]
  56. Guzewska, M.M.; Heifetz, Y.; Kaczmarek, M.M. Secretion patterns of extracellular vesicles during early pregnancy in the pig – in situ transmission electron microscopy study. In Proceedings of the 4th Winter Workshop of the Society for Biology of Reproduction, Zakopane, Poland, 13–15 February 2019. [Google Scholar]
  57. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell. Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
  58. 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]
  59. Hurley, J.H. The ESCRT complexes. Crit. Rev. Biochem. Mol. Biol. 2010, 45, 463–487. [Google Scholar] [CrossRef]
  60. Saftig, P.; Klumperman, J. Lysosome biogenesis and lysosomal membrane proteins: Trafficking meets function. Nat. Rev. Mol. Cell. Biol. 2009, 10, 623–635. [Google Scholar] [CrossRef] [PubMed]
  61. Yáñez-Mó, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles. 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Guzewska, M.M.; Heifetz, Y.; Kaczmarek, M.M. Expression and localization of extracellular vesicles biogenesis pathway elements in the porcine endometrium and conceptuses. In Proceedings of the 51st Annual Meeting of Society for the Study of Reproduction, New Orleans, LA, USA, 10–13 July 2018. [Google Scholar]
  63. McMahon, H.T.; Boucrot, E. Molecular mechanism and physiological functions of clathrin-mediated endocytosis. Nat. Rev. Mol. Cell. Biol. 2011, 12, 517–533. [Google Scholar] [CrossRef]
  64. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell. Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
  65. 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] [Green Version]
  66. 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]
  67. Tang, F.; Kaneda, M.; O’Carroll, D.; Hajkova, P.; Barton, S.C.; Sun, Y.A.; Lee, C.; Tarakhovsky, A.; Lao, K.; Surani, M.A. Maternal microRNAs are essential for mouse zygotic development. Genes Dev. 2007, 21, 644–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Nagaraja, A.K.; Andreu-Vieyra, C.; Franco, H.L.; Ma, L.; Chen, R.; Han, D.Y.; Zhu, H.; Agno, J.E.; Gunaratne, P.H.; DeMayo, F.J.; et al. Deletion of Dicer in somatic cells of the female reproductive tract causes sterility. Mol. Endocrinol. 2008, 22, 2336–2352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Fiedler, S.D.; Carletti, M.Z.; Hong, X.; Christenson, L.K. Hormonal regulation of MicroRNA expression in periovulatory mouse mural granulosa cells. Biol. Reprod. 2008, 79, 1030–1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. McBride, D.; Carré, W.; Sontakke, S.D.; Hogg, C.O.; Law, A.; Donadeu, F.X.; Clinton, M. Identification of miRNAs associated with the follicular- luteal transition in the ruminant ovary. Reproduction 2012, 144, 221–233. [Google Scholar] [CrossRef] [Green Version]
  71. Ma, T.; Jiang, H.; Gao, Y.; Zhao, Y.; Dai, L.; Xiong, Q.; Xu, Y.; Zhao, Z.; Zhang, J. Microarray analysis of differentially expressed microRNAs in non-regressed and regressed bovine corpus luteum tissue, microRNA-378 may suppress luteal cell apoptosis by targeting the interferon gamma receptor 1 gene. J. Appl. Gen. 2011, 52, 481–486. [Google Scholar] [CrossRef]
  72. Maalouf, S.W.; Liu, W.S.; Albert, I.; Pate, J.L. Regulating life or death: Potential role of microRNA in rescue of the corpus luteum. Mol. Cell. Endocrinol. 2014, 398, 78–88. [Google Scholar] [CrossRef]
  73. Gecaj, R.M.; Schanzenbach, C.I.; Kirchner, B.; Pfaffl, M.W.; Riedmaier, I.; Tweedie-Cullen, R.Y.; Berisha, B. The dynamics of microRNA transcriptome in bovine corpus luteum during its formation, function, and regression. Front. Genet. 2017, 8, 213. [Google Scholar] [CrossRef] [Green Version]
  74. Mohammed, B.T.; Sontakke, S.D.; Ioannidis, J.; Duncan, W.C.; Donadeu, F.X. The adequate corpus luteum: miR-96 promotes luteal cell survival and progesterone production. J. Clin. Endocrinol. Metab. 2017, 102, 2188–2198. [Google Scholar] [CrossRef] [Green Version]
  75. Diaz, F.J.; Wiltbank, M.C. Acquisition of luteolytic capacity involves differential regulation by prostaglandin F2a of genes involved in progesterone biosynthesis in the porcine corpus luteum. Domest. Anim. Endocrinol. 2005, 28, 172–189. [Google Scholar] [CrossRef]
  76. Diaz, F.J.; Luo, W.; Wiltbank, M.C. Effect of decreasing intraluteal progesterone on sensitivity of the early porcine corpus luteum to the luteolytic actions of prostaglandin F2alpha. Biol. Reprod. 2011, 841, 26–33. [Google Scholar] [CrossRef] [Green Version]
  77. Przygrodzka, E.; Witek, K.J.; Kaczmarek, M.M.; Andronowska, A.; Ziecik, A.J. Expression of factors associated with apoptosis in the porcine corpus luteum throughout the luteal phase of the estrous cycle and early pregnancy: Their possible involvement in acquisition of luteolytic sensitivity. Theriogenology 2015, 83, 535–545. [Google Scholar] [CrossRef] [PubMed]
  78. Przygrodzka, E.; Kaczmarek, M.M.; Kaczynski, P.; Ziecik, A.J. Steroid hormones, prostanoids, and angiogenic systems during rescue of the corpus luteum in pigs. Reproduction 2016, 15, 135–147. [Google Scholar] [CrossRef] [PubMed]
  79. Maalouf, S.W.; Smith, C.L.; Pate, J.L. Changes in MicroRNA expression during maturation of the bovine corpus luteum: Regulation of luteal cell proliferation and function by microRNA-34a. Biol. Reprod. 2016, 94, 71. [Google Scholar] [CrossRef] [PubMed]
  80. Stocco, C.O.; Zhong, L.; Sugimoto, Y.; Ichikawa, A.; Lau, L.F.; Gibori, G. Prostaglandin F2alpha-induced expression of 20alpha-hydroxysteroid dehydrogenase involves the transcription factor NUR77. J. Biol. Chem. 2000, 275, 37202–37211. [Google Scholar] [CrossRef] [Green Version]
  81. Stocco, C.O.; Lau, L.F.; Gibori, G. A calcium/calmodulin-dependent activation of ERK1/2 614 mediates JunD phosphorylation and induction of nur77 and 20alpha-hsd genes by prostaglandin F2alpha in 615 ovarian cells. J. Biol. Chem. 2002, 277, 3293–3302. [Google Scholar] [CrossRef] [Green Version]
  82. Atli, M.O.; Bender, R.W.; Mehta, V.; Bastos, M.R.; Luo, W.; Vezina, C.M.; Wiltbank, M.C. Patterns of gene expression in the bovine corpus luteum following repeated intrauterine infusions of low doses of prostaglandin F2alpha. Biol. Reprod. 2012, 86, 130. [Google Scholar] [CrossRef]
  83. Weber, J.A.; Baxter, D.H.; Zhang, S.; Huang, D.Y.; Huang, K.H.; Lee, M.J.; Galas, D.J.; Wang, K. The microRNA spectrum in 12 body fluids. Clin. Chem. 2010, 56, 1733–1741. [Google Scholar] [CrossRef]
  84. Wang, K.; Yuan, Y.; Cho, J.H.; McClarty, S.; Baxter, D.; Galas, D.J. Comparing the microRNA spectrum between serum and plasma. PLoS ONE 2012, 7, e41561. [Google Scholar] [CrossRef]
  85. 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 blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [Green Version]
  86. Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef] [Green Version]
  87. Vickers, K.C.; Palmisano, B.T.; Shoucri, B.M.; Shamburek, R.D.; Remaley, A.T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell. Biol. 2011, 13, 423–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Luo, S.S.; Ishibashi, O.; Ishikawa, G.; Ishikawa, T.; Katayama, A.; Mishima, T.; Takizawa, T.; Shigihara, T.; Goto, T.; Izumi, A.; et al. Human villous trophoblasts express and secrete placenta-specific microRNAs into maternal circulation via exosomes. Biol. Reprod. 2009, 81, 717–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Mouillet, J.F.; Ouyang, Y.; Coyne, C.B.; Sadovsky, Y. MicroRNAs in placental health and disease. Am. J. Obstet. Gynecol. 2015, 213, S163–S172. [Google Scholar] [CrossRef] [Green Version]
  90. Ioannidis, J.; Donadeu, F.X. Circulating miRNA signatures of early pregnancy in cattle. BMC Genom. 2016, 17, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Guelfi, G.; Stefanetti, V.; De Luca, S.; Giontella, A.; Barile, V.L.; Barbato, O. Serum microRNAs in buffalo cows: Potential biomarkers of pregnancy. Res. Vet. Sci. 2017, 115, 294–300. [Google Scholar] [CrossRef]
  92. Ioannidis, J.; Donadeu, F.X. Changes in circulating microRNA levels can be identified as early as day 8 of pregnancy in cattle. PLoS ONE 2017, 12, e0174892. [Google Scholar] [CrossRef] [Green Version]
  93. 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]
  94. Klohonatz, K.M.; Cameron, A.D.; Hergenreder, J.R.; da Silveira, J.C.; Belk, A.D.; Veeramachaneni, D.N.; Bouma, G.J.; Bruemmer, J.E. Circulating miRNAs as potential alternative cell signaling associated with maternal recognition of pregnancy in the mare. Biol. Reprod. 2016, 95, 124. [Google Scholar] [CrossRef]
  95. Cortez, M.A.; Bueso-Ramos, C.; Ferdin, J.; Lopez-Berestein, G.; Sood, A.; Calin, G.A. MicroRNAs in body fluids—the mix of hormones and biomarkers. Nat. Rev. Clin. Oncol. 2011, 8, 467–477. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Uptake of EVs via clathrin-maintained endocytosis by trophoblast cell on day 16 of pregnancy. Two types of EVs internalized by endocytosis are located in the extracellular space. Orange–large vesicles ectosomes, and yellow–small exosomes. Clathrin-coated pits are marked by black dashed line.
Figure 1. Uptake of EVs via clathrin-maintained endocytosis by trophoblast cell on day 16 of pregnancy. Two types of EVs internalized by endocytosis are located in the extracellular space. Orange–large vesicles ectosomes, and yellow–small exosomes. Clathrin-coated pits are marked by black dashed line.
Ijms 21 02229 g001
Figure 2. Proposed model of E2-mediated regulation of miRNA expression in luteal tissue during early pregnancy in pigs. E2 enters the luteal cell and binds with its genomic receptor, ERα, located in the cytoplasm. It leads to formation of ERα homodimers and their translocation to the nucleus. In the nucleus, ERα homodimers bind to (1) an estrogen-responsive element (ERE) or (2) transcription factor SP1, which binds to transcriptional factor responsive elements (TFRE) in the promoter region of miR-99b cluster. As a result, there is an increase in the expression of miRNAs belonging to miR-99b cluster (miR-99b, miR-125a, and let-7e), which can target mRNA involved in luteolysis including NR4A1, nuclear receptor subfamily 4 group A member 1; EDN1, endothelin 1; FOS, fos proto-oncogene; JUN, jun proto-oncogene. That mechanisms support the function of luteal cells and allow continuation of progesterone production required for pregnancy establishment and maintenance.
Figure 2. Proposed model of E2-mediated regulation of miRNA expression in luteal tissue during early pregnancy in pigs. E2 enters the luteal cell and binds with its genomic receptor, ERα, located in the cytoplasm. It leads to formation of ERα homodimers and their translocation to the nucleus. In the nucleus, ERα homodimers bind to (1) an estrogen-responsive element (ERE) or (2) transcription factor SP1, which binds to transcriptional factor responsive elements (TFRE) in the promoter region of miR-99b cluster. As a result, there is an increase in the expression of miRNAs belonging to miR-99b cluster (miR-99b, miR-125a, and let-7e), which can target mRNA involved in luteolysis including NR4A1, nuclear receptor subfamily 4 group A member 1; EDN1, endothelin 1; FOS, fos proto-oncogene; JUN, jun proto-oncogene. That mechanisms support the function of luteal cells and allow continuation of progesterone production required for pregnancy establishment and maintenance.
Ijms 21 02229 g002
Figure 3. Peri-implantation events are governed by various biological molecules, including miRNAs produced by the endometrium and conceptus in pigs. miRNAs carried by EVs participate in reciprocal communication between the conceptus and endometrium, regulating the expression of genes involved in several pathways crucial for successful implantation. Tr, trophoblast; LE, luminal epithelium; GE, glandular epithelium; ST, stroma; NK cells, natural killer cells. Scale bar, 200 μm.
Figure 3. Peri-implantation events are governed by various biological molecules, including miRNAs produced by the endometrium and conceptus in pigs. miRNAs carried by EVs participate in reciprocal communication between the conceptus and endometrium, regulating the expression of genes involved in several pathways crucial for successful implantation. Tr, trophoblast; LE, luminal epithelium; GE, glandular epithelium; ST, stroma; NK cells, natural killer cells. Scale bar, 200 μm.
Ijms 21 02229 g003
Table 1. Examples of microRNAs (miRNAs) detected in pregnant pigs.
Table 1. Examples of microRNAs (miRNAs) detected in pregnant pigs.
Source of MiRNAsExamples of Detected MiRNAsRef.
Embryo8 cellmiR-17, -125a-5p, -125b, -128, -205[17]
blastocystmiR -92a, -129-5p, -205, -210, -302a[17]
day 10miR-371-3p, -455, -449a, -634, -940, -1193-3p[22]
miR-7, -302b, -302d, -371-5p, -378[38]
day 12miR-96, -138, -152, -483-5p, -765[22]
miR-301, -467, -4057[34]
day 14miR-10a, -21, -23b, -200a, -301a, -574[34]
day 16 (trophoblast)miR-26a, -27a, -148a[22]
day 20 (trophoblast)miR-125b, -199a-3p, -199a-5p[22]
miR-15b-5p, -18a, -20a-5p, -126-5p, -155-5p, -221-5p[32]
day 25 (placenta)miR-17, -18a, -19a, -20a, -92a[36]
day 30 (placenta)miR-17, -106a, -107, -345-5p, -615-3p[35]
day 50 (trophoblast/placenta)let-7f-5p, miR-150, -221-5p,-222,[32]
let-7a/b/c/d/e/f/i, -23a, -24, -27a/b, -29a,
-30a/e, -141, -205
[36]
Endometriumday 12let-7f/g/i, miR-10b/d, -143-3p[38]
day 15let-7b/c/i, miR-125a-3p, -135a*, -140-3p, -149,
-181c/d, -200c, -361, -494, -542-5p
[37]
day 16miR-23b, -127, -411, -449a[23]
day 20miR-1, -10b, -27a, -30d -101-1, -126, -143, -146b,
-193, -193b-3p, -205, -574
[23]
miR-21, -22-3p, -27a -29a, -30b-5p, -30d, -30e-5p, -149, -183, -191, -205, -296, -323, -362, -432-3p,-503, -574, -4335, -4339[33]
day 26miR-15b, -17-5p, -18a, 20a, 30a-3p/5p,
-92b, -106a, -126, -132, -221, -222
[37]
day 50miR-30c/e, -215, -411, -487b[37]
EVs1ULFs2 from gestational
day 14 and 16
miR-26a, -125b[22]
CM3 of porcine trophectoderm cellsmiR-15b, -16, -17-5p, -20a, -126-5p, -150,
-155-5p, -221-5p
[39]
OvaryCL from gestational
day 14
miR-21a-3p, -345-3p, -371-5p, -4334-5p,
-9788-3p, -9840-3p, -9850-5p
[40]
Serumgestational
day 16 and 20
let-7a/b/c/f, miR-10a, -23b, -26a, -30b-5p,
-125b, -143-3p
[41,42]
1 EVs—extracellular vesicles; 2 ULFs—uterine luminal flushings; 3 CM—conditioned media

Share and Cite

MDPI and ACS Style

Kaczmarek, M.M.; Najmula, J.; Guzewska, M.M.; Przygrodzka, E. MiRNAs in the Peri-Implantation Period: Contribution to Embryo–Maternal Communication in Pigs. Int. J. Mol. Sci. 2020, 21, 2229. https://doi.org/10.3390/ijms21062229

AMA Style

Kaczmarek MM, Najmula J, Guzewska MM, Przygrodzka E. MiRNAs in the Peri-Implantation Period: Contribution to Embryo–Maternal Communication in Pigs. International Journal of Molecular Sciences. 2020; 21(6):2229. https://doi.org/10.3390/ijms21062229

Chicago/Turabian Style

Kaczmarek, Monika M., Joanna Najmula, Maria M. Guzewska, and Emilia Przygrodzka. 2020. "MiRNAs in the Peri-Implantation Period: Contribution to Embryo–Maternal Communication in Pigs" International Journal of Molecular Sciences 21, no. 6: 2229. https://doi.org/10.3390/ijms21062229

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