Next Article in Journal
Tracking Changes in the Spring Barley Gene Pool in Poland during 120 Years of Breeding
Previous Article in Journal
Comparative Efficacy and Safety of P2Y12 Inhibitor Monotherapy and Dual Antiplatelet Therapy in Patients with and without Diabetes Mellitus Undergoing Percutaneous Coronary Intervention
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circular RNAs in Pregnancy and the Placenta

by
Anya L. Arthurs
*,
Tanja Jankovic-Karasoulos
,
Melanie D. Smith
and
Claire T. Roberts
*
Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(9), 4551; https://doi.org/10.3390/ijms23094551
Submission received: 23 March 2022 / Revised: 16 April 2022 / Accepted: 19 April 2022 / Published: 20 April 2022
(This article belongs to the Section Molecular Immunology)

Abstract

:
The emerging field of circular RNAs (circRNAs) has identified their novel roles in the development and function of many cancers and inspired the interest of many researchers. circRNAs are also found throughout the healthy body, as well as in other pathological states, but while research into the function and abundance of circRNAs has progressed, our overall understanding of these molecules remains primitive. Importantly, recent studies are elucidating new roles for circRNAs in pregnancy, particularly in the placenta. Given that many of the genes responsible for circRNA production in cancer are also highly expressed in the placenta, it is likely that the same genes act in the production of circRNAs in the placenta. Furthermore, placental development can be referred to as ‘controlled cancer’, as it shares many key signalling pathways and hallmarks with tumour growth and metastasis. Hence, the roles of circRNAs in this field are important to study with respect to pregnancy success but also may provide novel insights for cancer progression. This review illuminates the known roles of circRNAs in pregnancy and the placenta, as well as demonstrating differential placental expressions of circRNAs between complicated and uncomplicated pregnancies.

1. Introduction

The placenta, a product of conception with a transient existence, uniquely supports pregnancy. It plays a critical role in nutrient, waste and gas exchange between the mother and fetus. Correct placentation underpins fetal development, as well as coordinating maternal adaptations to pregnancy to maintain maternal and fetal health. In pregnancy complications characterised by aberrant placentation such as preeclampsia (PE) [1] and intrauterine growth restriction [2], there is an altered placental transcriptome. Emerging evidence demonstrates the roles of novel RNA species in pregnancy complications, particularly circular RNAs (circRNAs).
The first identified circRNA, the hepatitis D viroid, was reported in 1977 [3]. Following this, circRNAs were found in mammalian cells using electron microscopy in 1979 [4]. Initially, due to their low abundance, they were disregarded as products of misplicing. Then, in 1991, circular transcripts were found in a variety of normal and neoplastic human cells [5]. As technology improved, primarily in sequencing capabilities and bioinformatics, so has the study of the structure and functionality of circRNAs [6].
Many circRNAs have evaded detection until now for two main reasons. Unlike other small RNAs, circRNAs are not able to be easily separated from mRNA through size fractionation or electrophoretic mobility as often they differ from their linear form only in circular structure. They are also easily destroyed by molecular techniques requiring amplification or fractionation due to their circular form and, as they lack polyadenylation, they are often discarded when analysing sequencing data [7]. These covalently closed circular RNA structures remain enigmatic, with a plethora of reported functions and methods of biogenesis. This review will detail what is currently known about circRNAs, their implications for placental development and function, and their broader consequences for pregnancy.

2. circRNA Biogenesis

circRNAs are produced through backsplicing, a process in which the 3′ end of a downstream exon is spliced and covalently linked with the 5′ end of an upstream exon [8,9,10]. This often leaves behind a transcript which becomes an alternatively spliced linear RNA product with skipped exons (Figure 1) [11]. The end circRNA product is devoid of 5′ capping and 3′ polyadenylation, and is consequently resistant to exonuclease activity [12]. It is possible for circRNAs to consist of one or more exons, sometimes including introns [termed exonic intronic circRNAs (EIcircRNAs)], or even introns only [termed circular intronic RNAs (ciRNAs)]. Exonic circRNAs make up approximately 80% of circRNA transcripts and are mainly located in the cytoplasm, whereas EIcircRNAs and ciRNAs tend to be located in the nucleus and regulate their cognate linear transcripts [13].
It has been shown that the biogenesis of circRNAs, while via backsplicing, still involves canonical splicing signals and spliceosomal mechanisms [12]. The experimental use of isoginkgetin, a splicing inhibitor, inhibits the formation of circRNAs, as well as linear RNAs [14,15]. Moreover, mutations in the canonical splicing sites of exons inhibit circularisation and circRNA biogenesis [16,17,18]. The biogenesis of circRNAs is in constant competition with linear RNA production through canonical splicing machinery [15]. It has been shown that the elongation velocity of RNA polymerase II positively correlates with backsplicing efficiency [19]. This has been corroborated by several studies in which mutations in the RNA polymerase II large subunit significantly reduced RNA pol II elongation velocity, and thus, backsplicing efficiency and circRNA production [20,21,22].
There are several ways in which circRNAs can be produced (Figure 2). Complementary base-pairing (Figure 2A) occurs when complementary inverted sequences in introns flanking backsplice junctions facilitate circularisation by base-pairing to form a stem-loop-like structure which can then be cleaved to form a circRNA. This structure promotes spatial reduction in splice signals required for backsplicing and thus contributes to RNA circularization [23,24]. Specifically, Jeck et al. [25] first reported on the importance of inverted ALU repeat elements in backsplice-flanking introns in facilitating circRNA biogenesis. ALU repeats are short nucleotide sequence repeats that comprise approximately 11% of the genome and are primate-specific [26]. These inverted ALU repeats are five times more enriched in sites of human exonic circRNAs formation. There are also examples of exonic circRNAs where the entire gene is circularised and no upstream or downstream exons are leftover for alternatively spliced transcript production, such as SRY, the male sex-determining gene found on the Y chromosome, which abundantly produces circRNAs [27].
Certain RNA binding proteins (RBPs) are also able to facilitate RNA circularisation (Figure 2B). For example, the RBP Quaking (QKI), which is highly expressed in the placenta, aids the biogenesis of circRNAs which are involved in epithelial-mesenchymal transition (EMT), a process common to placental development and many cancers. Furthermore, QKI knockdown subsequently inhibits the production of EMT-related circRNAs [18]. However, for correct functioning QKI requires the assistance of binding sites in introns flanking the exons to facilitate circRNA biogenesis [18]. Alternatively, the RBP Muscleblind (MBL), which is also highly expressed in the placenta, facilitates the biogenesis of the circRNA (circMbl) from its own cognate RNA by binding to specific MBL conserved sites in flanking introns [15].
circRNAs can also be formed from RNA lariats (lasso-shaped by-products of RNA splicing), termed circular intronic RNAs (ciRNAs). Distinct from exonic circRNAs, which feature a 3′-5′ carbon linkage at the splicing branchpoint, lariat RNAs feature 2′-5′ linkages [23]. They can be formed utilising a consensus motif with a GU-rich region, located near the 5′ splicing point, and a C-rich region, near the branchpoint site in ciRNA-producing introns, which allow for intron lariat escape from debranching. These regions then facilitate the circularisation of this intron [28,29] (Figure 2C) and the 3′ ‘tail’ downstream from the branch point is trimmed to stabilise the ciRNA and protect from exonucleases. This motif is not enriched in regular introns [23] and has been suggested as an essential RNA element to expedite intron lariat escape from debranching.
The lariat-driven model of circularisation (Figure 2D) can encompass a variety of the above techniques for circRNA biogenesis. Middle exons of a linear transcript are ‘skipped’ to allow an upstream 3′ splice donor to covalently bond to a downstream 5′ splice acceptor. The spliceosome then removes the introns to form the final circRNA product.

3. circRNA Function

There are several functions for circRNAs that have been identified to date. A small number of circRNAs are able to be translated (Figure 3A) (e.g., the Hepatitis δ agent, a circular RNA satellite virus of the Hepatitis B virus [30]) while engineered circRNAs can undergo translation if an internal ribosomal entry site (IRES) is included in the design [7]. However, the majority of circRNAs appear to be non-coding.
Some specific, highly expressed circRNAs function as miRNA sponges (Figure 3B). The exonic circRNAs from CDR1as [31], cerebellum-related antigen 1, and SRY [12], the testis-determining factor, have been shown to bind miRNAs without degrading them, inhibiting their function. Each of these circRNAs also has multiple miRNA binding sites in its sequence. The circRNA for CDR1as has 74 confirmed sites for miR-7 binding, as well as being densely seeded with Argonaute protein binding sites which allow for Argonaute–miRNA complexes to bind. The circRNA for SRY has 16 binding sites for miR-138 and coprecipitates with Argonaute 2. However, the concept that circRNAs act as miRNA sponges has recently been debated.
Whilst it is true that some circRNAs function efficiently as miRNA sponges, such as ciRS-7 [12], the notion of circRNAs functioning as sponges has been questioned due to the stoichiometric ratio of circRNA to miRNA molecules within the cell [32]. Given that the majority of circRNAs are produced at less than 2.5 copies per cell [18], it is improbable that they are able to significantly regulate the expression of miRNAs, which are often produced at 900–80,000 copies per cell [33]. The potential for circRNAs to mediate miRNA expression is likely to be reserved only for circRNAs with unusually high expression within cells, and multiple miRNA binding sites per molecule. Thus, new studies to examine the potential function of circRNAs as miRNA sponges may need to seek further validation through experiments that involve more than dual luciferase assays. However, this is not to suggest that many circRNAs do not have important cellular functions. As the majority of circRNAs are produced at ~2.5 copies per cell, this indicates an approximate 1:1 ratio with the DNA transcripts in each cell. Indeed, interaction with DNA is another function of circRNAs that has important implications in molecular biology (this is explored further below).
circRNAs can also function as transcriptional regulators, termed “mRNA traps” (Figure 3C). One example of this is the exonic circRNA produced from the Fmn (flavin mononucleotide) gene in mice, which is proposed to sequester the translation start site on the mRNA, reducing protein synthesis [34]. circRNAs can also bind proteins (Figure 3D), as previously mentioned, circMbl can sequester the Muscleblind RBP [15]. Furthermore, circANRIL, a circRNA in the antisense non-coding RNA in the INK4 locus (ANRIL) long non-coding RNA, regulates the maturation of precursor ribosomal RNA, therefore controlling ribosome biogenesis [15]. circRNAs have also been shown to facilitate the phosphorylation, ubiquitylation and acetylation [15] of proteins (Figure 3E), and participate as structural components of protein complexes [15]. Importantly, circRNAs have been shown to bind to, and facilitate breakages in DNA (see below) (Figure 3F). circRNAs have also been reported to recruit proteins to specific subcellular loci [15] and influence host transcript promoter regions (Figure 3G). With their many attributed functions, it is no surprise that circRNAs have been implicated to play a role in many pathophysiological and physiological states; this review will focus on their role in pregnancy.

4. The Role of circRNAs in Pregnancy

circRNAs are expressed throughout reproductive tissues in healthy pregnancy and are differentially expressed between healthy and complicated pregnancy. However, the question of whether this is cause or effect requires further research. Studies that have examined circRNAs expressed in reproductive tissues and detected in maternal serum in pregnancy are limited and summarised in Table 1.
In animals, research conducted in murine models has described circRNA profiles in oocytes and pre-implantation embryos [35] and in both implantation and inter-implantation sites in the endometrium [41]. The sizable differences in the profiles between these cells and tissues indicate that circRNAs play a role in the reproductive process. Interestingly, one study was completed in both in vitro cell lines and in vivo rat experiments to demonstrate the effect of circSFXN1 (sideroflexin 1) in PE pathology [53]. sFLT1-expressing adenovirus injections into rats induced a PE-like phenotype, which was abated by treatments with si-circSFXN1. This clearly demonstrates the pathological potential for aberrant circRNA expression. Another study examined atretic follicles in porcine ovaries, determining that a circSLC41A1-miR-9820-5p-SRSF1 axis regulates follicular granulosa cell apoptosis [85].
In humans, circRNAs have been profiled in granulosa cells in ovarian follicles [40], placenta [38,39,42,44,45,46,47,51,52,53,54,55,57,58,59,66,67,68,70,72,75,76,78,79,80,81,82,83,84], a multitude of different fetal tissues [36] and maternal blood [37,43,45,49,50,52,71,73,74,77,83], as well as exosomes isolated from umbilical cord blood [48,69]. Interestingly, one study confirmed that pregnancy-specific circRNAs were able to be detected in first-trimester platelets [49]. Many of these studies demonstrate differential circRNA expression profiles for disease states compared with an uncomplicated pregnancy, particularly comparisons in circRNA expression between PE, or gestational diabetes mellitus (GDM) and an uncomplicated pregnancy control. Some studies then went on to suggest particular circRNAs with biomarker potential for the pregnancy complication. Better studies followed this assertion by then performing functional studies to elucidate mechanisms of action for circRNAs of interest, utilising cell lines with circRNA overexpression or knockdown (data are presented in Table 1).

4.1. circRNAs in Preeclampsia

In placentae from women with PE, circ_0001438 [57], circ_0001687 [84], circ_0001855 [43], circ_0004904 [61], circ_0008726 [78], circ_0011460 [66], circ_0026552 [70], circ_0036877 [45], circ_0037078 [75], circ_0085296 [55], circ_0111277 [67], circ_101222 [37], circ_3286 [44], circBRAP [83], circLRRK1 [59], circSFXN1 [53], circTNRC18 [46] and circZDHHC20 [54] were elevated compared with uncomplicated pregnancy controls. A subset of these (circ_0001438, circ_0004904, circ_0008726, circ_0011460, circ_0026552, circ_0037078, circ_0085296, circ_0111277, circ_3286, circBRAP, circLRRK1, circSFXN1, circTNRC18 and circZDHHC20) when overexpressed in vitro resulted in decreased cell proliferation, migration, invasion or angiogenesis, or a combination of these effects. In contrast, circ_0001513 [84], circ_0007121 [68], circ_0017068 [82], circ_0032962 [76], circ_0051326 [77], circHIPK3 [80], circPAPPA [81], circ_PAPPA2 [79] and circUBAP2 [56] were decreased in PE placentae. A subset of these (circ_0007121, circ_0017068, circ_0032962, circHIPK3, circPAPPA and circUBAP2) when expressed in vitro promoted cell proliferation, migration, invasion or angiogenesis, or a combination of these effects. Many circRNAs studied were suggested to perform these functions through miRNA sponging but, as previously mentioned, the physiological impact of lowly expressed circRNAs sponging highly abundant miRNAs is debatable.

4.2. circRNAs in Gestational Diabetes Mellitus

Studies on placental circRNA expression in GDM focused mainly on profiling differences between GDM and uncomplicated pregnancies. In one study, first and early second-trimester maternal blood samples were collected to compare circRNA differential expression. These measures were then used to determine possible circRNA predictors for GDM development [50]. Other studies showed that circ_0008285 [60], circ_0026497 [71], circ_0039480 [71], circ-PNPT1 [63] and circVEGFC [74] were elevated in maternal plasma and whole blood from women with GDM. In vitro experiments using high glucose media for HTR-8/SVneo cell culture promoted proliferation and migration, which was reversed with circ_0008285 knockdown. Similarly, high glucose-induced arrest of cell viability and migration was reversed upon circ-PNPT1 knockdown. High levels of circVEGFC occurred with higher incidence rates of fetal malformation and hypertension. circ_0074673 [69] was upregulated in exosomes isolated from umbilical cord blood of GDM cases.
In contrast, other studies showed that circ_0001173 [60], circ_0005243 [52] and circ_102682 [73] were downregulated in placentae and maternal plasma from women with GDM. In vitro knockdown of circ_0005243 in HTR-8/SVneo trophoblast cells suppressed cell proliferation and migration, while circ_0001173 levels were positively correlated with glycated haemoglobin.

4.3. circRNAs in Other Pregnancy Complications

Other pregnancy complications have also been briefly studied with respect to circRNAs. One study reported almost 600 differentially expressed circRNAs in placentae from women with recurrent spontaneous abortion (RSA) compared with uncomplicated pregnancy [39]. Another study observed that circ_0050703 was downregulated in the placental villous tissue of patients with unexplained RSA (URSA), and circ_0050703 silencing in vivo reduced the number of successfully implanted embryos [64]. A circFOXP1/miR-143-3p/S100A11 axis was suggested in the RSA placentae [72]. Furthermore, circ-SETD2 was implicated in placental growth, with elevated circ-SETD2 in placentae of patients with fetal macrosomia [51]. In vivo overexpression experiments in HTR-8/SVneo cells showed increased cell proliferation and invasion. A circ_0074371/miR-582-3p/LRP6 axis was suggested in the context of fetal growth restriction [65]. Finally, granulosa cells from non-pregnant advanced age (≥38 years) compared with young age (≤30 years) women determined different circRNAs expression profiles depending on maternal age [40]. Whilst the number of studies into circRNAs in pregnancy is low, clearly circRNAs play many roles in pregnancy health, waiting to be discovered.

4.4. Limitations of circRNA Research

Research surrounding circRNAs in pregnancy is certainly still in its infancy. Several studies (Table 1) reported only the results of their profiling without any qPCR validation in independent samples. Whilst these data could be useful to other researchers, many of these profiling techniques have now been superseded with novel technologies. Methods such as RPAD [86], along with the use of Li+ ions in reaction buffers [87], in RNA-sequencing (RNA-seq) are proving much more reliable than the outdated circRNA arrays. circRNA detection through RNA-seq can be accurate assuming that one of these above methods is employed to prepare the samples.
Importantly, many of the studies described in this review lacked RNase R enrichment prior to sequencing. The addition of this exonuclease to a sample results in the digestion of all linear RNAs present, leaving an enriched population of circRNA transcripts. Not using this treatment prior to sequencing means that the depth of sequencing for circRNAs will be limited given that the sample is still composed primarily of linear RNA products. This likely results in the detection of only the most highly expressed circRNAs, leaving many transcripts undetected.
Finally, in studies using delivered placentae, authors should be cautious to declare specific circRNAs as causes of a pregnancy state. Given that these placentae have been collected after birth, without further mechanistic studies, it is unclear whether circRNAs in disease states are causal or an effect. However, these few studies provide enticing evidence to inspire further research into circRNAs in pregnancy and placental growth.

5. The Potential Importance of circRNAs in the Placenta: What We Can Apply from Our Knowledge of Cancer

The placenta can be considered a ‘controlled cancer’, as there are many parallels that have been previously drawn between placental development and cancer metastasis [88,89,90]. Some basic principles of cancer progression include tumour growth and tissue invasiveness (through epithelial to mesenchymal transition) [91], immune evasion and stimulation of angiogenesis [92], all of which are essential for successful placentation [93,94]. It is therefore unsurprising that many of the key molecular pathways are common to both placental development and cancer (Table 2). Importantly, the extensive research demand in the field of cancer has yielded a wealth of information about the molecular biology of cancers that can also be applied to the study of the placenta due to their many similarities.
Extensive mRNA profiling has been undertaken in placentae from the first trimester, second trimester and term [120,121,122,123]. Utilising this dataset shows that many of the genes responsible for circRNA production in cancer are also highly expressed in the placenta [124]. Hence, it is likely that circRNAs are also produced from these genes in the placenta. For example; the circRNA produced from MYLK has been shown to interfere with VEGFA signalling in bladder cancer [125]. The importance of the VEGF signalling pathway is well established in placentation [107], being most important for early angiogenesis and maintaining vascular health in the mother. Conversely, MYLK expression in the placenta increases across gestation. It is highly likely that circRNAs from the MYLK gene are also produced in the placenta and could impact the VEGF signalling pathway. There appear to be endless examples of genes which are known to produce circRNAs in cancer that are relevant to, and highly expressed in, placental development. We are currently profiling several of these circRNAs in the placenta and examining their roles in its development.
Importantly, circRNAs are not only able to affect development through their interactions with other molecules but they can also facilitate genomic instability through translocations. Rapid proliferation and consequent replication stress are common [126], both in cancer and in the placenta, resulting in possible DNA damage. Evidence has been provided for R-loop formation in plants, where a circRNA forms an RNA:DNA hybrid with its cognate DNA locus, stalling transcription and resulting in DNA breaks [127]. This was also shown to coincide with the recruitment of splicing factors, as well as alternative splicing. This genomic manipulation by circRNAs is likely to also occur in the eukaryotic tissues, although this is yet to be confirmed. If this is the case, dysregulation of circRNAs, particularly in early gestation, could result in genomic alterations that could affect both placental and fetal development and pregnancy health in general.
circRNAs have been shown to accumulate in a number of different tissues over time [128,129] and have been suggested to be a marker of tissue ageing. As the placenta is also known to undergo ageing [130], it is possible that circRNA accumulation in the placenta could occur. The implications of circRNA accumulation are still not well understood, but if these circRNAs continue to exert their functions as they accumulate this could lead to exaggerated circRNA action in the tissue.

6. Conclusions

Understanding the functions of circRNAs, particularly their involvement in placental development and pregnancy health, is in its infancy. However, these unique molecules are evidently the result of careful regulation, with multiple roles in physiological and pathophysiological conditions. Evidence that circRNAs may be involved in regulating placental development, and that differential circRNA profiles are found between healthy and complicated pregnancies, provides an imperative for further research.

Author Contributions

Conceptualization, A.L.A.; writing—original draft preparation, A.L.A.; writing—review and editing, T.J.-K., M.D.S. and C.T.R.; visualization, A.L.A.; supervision, C.T.R.; funding acquisition, C.T.R. All authors have read and agreed to the published version of the manuscript.

Funding

C.T.R. is funded by a National Health and Medical Research Council Investigator Grant (GNT1174971) and a Matthew Flinders Professorial Fellowship from Flinders University, Australia. A.L.A. is supported by funding from the Flinders Foundation and Flinders University.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Figures were made using Biorender.com, accessed on 5 May 2021.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sõber, S.; Reiman, M.; Kikas, T.; Rull, K.; Inno, R.; Vaas, P.; Teesalu, P.; Marti, J.M.L.M.; Pirkko, M.; Laan, M. Extensive shift in placental transcriptome profile in preeclampsia and placental origin of adverse pregnancy outcomes. Sci. Rep. 2015, 5, 13336. [Google Scholar] [CrossRef] [PubMed]
  2. Majewska, M.; Lipka, A.; Paukszto, L.; Jastrzebski, J.; Szeszko, K.; Gowkielewicz, M.; Lepiarczyk, E.; Jozwik, M.; Majewski, M. Placenta Transcriptome Profiling in Intrauterine Growth Restriction (IUGR). Int. J. Mol. Sci. 2019, 20, 1510. [Google Scholar] [CrossRef] [PubMed]
  3. Rizzetto, M.; Canese, M.; Arico, S.; Crivelli, O.; Trepo, C.; Bonino, F.; Verme, G. Immunofluorescence detection of new antigen-antibody system (delta/antidelta) associated to hepatitis B virus in liver and in serum of HBsAg carriers. Gut 1977, 18, 997–1003. [Google Scholar] [CrossRef] [PubMed]
  4. Hsu, M.; Coca-Prados, M. Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature 1979, 280, 339–340. [Google Scholar] [CrossRef] [PubMed]
  5. Nigro, J.; Cho, K.; Fearon, E.; Kern, S.; Ruppert, J.; Oliner, J.; Kinzler, K.; Vogelstein, B. Scrambled exons. Cell 1991, 64, 607–613. [Google Scholar] [CrossRef]
  6. Ozsolak, F.; Milos, P. RNA sequencing: Advances, challenges and opportunities. Nat. Rev. Genet. 2011, 12, 87–98. [Google Scholar] [CrossRef]
  7. Jeck, W.; Sharpless, N. Detecting and characterizing circular RNAs. Nat. Biotechnol. 2014, 32, 423–461. [Google Scholar] [CrossRef]
  8. Salzman, J.; Gawad, C.; Wang, P.; Lacayo, N.; Brown, P. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS ONE 2012, 7, e30733. [Google Scholar] [CrossRef]
  9. Guo, J.; Agarwal, V.; Guo, H.; Bartel, D. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014, 15, 409. [Google Scholar] [CrossRef]
  10. Memczak, S.; Jens, M.; Elefsinioti, A.; Torti, F.; Krueger, J.; Rybak, A.; Maier, L.; Mackowiak, S.; Gregersen, L.; Munschauer, M.; et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 2013, 495, 333–338. [Google Scholar] [CrossRef]
  11. Chen, L.; Yang, L. Regulation of circRNA biogenesis. RNA Biol. 2015, 12, 381–388. [Google Scholar] [CrossRef] [PubMed]
  12. Hansen, T.; Jensen, T.; Clausen, B.; Bramsen, J.; Finsen, B.; Damgaard, C. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388. [Google Scholar] [CrossRef] [PubMed]
  13. Floris, G.; Zhang, L.; Follesa, P.; Sun, T. Regulatory Role of Circular RNAs and Neurological Disorders. Mol. Neurobiol. 2017, 54, 5156–5165. [Google Scholar] [CrossRef]
  14. Starke, S.; Jost, I.; Rossbach, O.; Schneider, T.; Schreiner, S.; Hung, L.; Bindereif, A. Exon circularization requires canonical splice signals. Cell Rep. 2015, 10, 103–111. [Google Scholar] [CrossRef] [PubMed]
  15. Ashwal-Fluss, R.; Meyer, M.; Pamudurti, N.; Ivanov, A.; Bartok, O.; Hanan, M.; Evantal, N.; Memczak, S.; Rajewsky, N.; Kadener, S. circRNA biogenesis competes with pre-mRNA splicing. Mol. Cell 2014, 56, 55–56. [Google Scholar] [CrossRef] [PubMed]
  16. Quan, G.; Li, J. Circular RNAs: Biogenesis, expression and their potential roles in reproduction. J. Ovarian Res. 2018, 11, 9. [Google Scholar] [CrossRef]
  17. Chen, I.; Chen, C.; Chuang, T. Biogenesis, identification, and function of exonic circular RNAs. Wiley Interdiscip. Rev. RNA 2015, 6, 563–579. [Google Scholar] [CrossRef]
  18. Conn, S.J.; Pillman, K.A.; Toubia, J.; Conn, V.M.; Salmanidis, M.; Phillips, C.A.; Roslan, S.S.; Andreas, W.; Gregory, P.A.; Goodall, G.J. The RNA binding protein quaking regulates formation of circRNAs. Cell 2015, 160, 1125–1134. [Google Scholar] [CrossRef]
  19. Ebbesen, K.; Hansen, T.; Kjems, J. Insights into circular RNA biology. RNA Biol. 2017, 14, 1035–1045. [Google Scholar] [CrossRef]
  20. De La Mata, M.; Alonso, C.; Kadener, S.; Fededa, J.; Blaustein, M.; Pelisch, F.; Cramer, P.; Bentley, D.; Kornblihtt, A. A slow RNA polymerase II affects alternative splicing in vivo. Mol. Cell 2003, 2, 525–532. [Google Scholar] [CrossRef]
  21. Ip, J.; Schmidt, D.; Pan, Q.; Ramani, A.; Fraser, A.; Odom, D.; Blencowe, B. Global impact of RNA polymerase II elongation inhibition on alternative splicing regulation. Genome Res. 2011, 21, 390–401. [Google Scholar] [CrossRef] [PubMed]
  22. Khodor, Y.; Rodriguez, J.; Abruzzi, K.; Tang, C.; Marr, M.; Rosbash, M. Nascent-seq indicates widespread cotranscriptional pre-mRNA splicing in drosophila. Genes Dev. 2011, 23, 2502–2512. [Google Scholar] [CrossRef]
  23. Zhang, Y.; Zhang, X.O.; Chen, T.; Xiang, J.F.; Yin, Q.F.; Xing, Y.H.; Zhu, S.; Yang, L.; Chen, L.L. Circular intronic long noncoding RNAs. Mol. Cell 2013, 51, 792–806. [Google Scholar] [CrossRef] [PubMed]
  24. Ivanov, A.; Memczak, S.; Wyler, E.; Torti, F.; Porath, H.; Orejuela, M.; Piechotta, M.; Levanon, E.; Landthaler, M.; Dieterich, C.; et al. Analysis of intron sequences reveals hallmarks of circular RNA biogenesis in animals. Cell Rep. 2015, 10, 170–177. [Google Scholar] [CrossRef] [PubMed]
  25. Jeck, W.; Sorrentino, J.; Wang, K.; Slevin, M.; Burd, C.; Liu, J. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 2013, 19, 141–157. [Google Scholar] [CrossRef] [PubMed]
  26. Lander, E.; Linton, L.; Birren, B.; Nusbaum, C.; Zody, M.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921. [Google Scholar] [CrossRef]
  27. Capel, B.; Swain, A.; Nicolis, S.; Hacker, A.; Walter, M.; Koopman, P.; Goodfellow, P.; Lovell-Badge, R. Circular transcripts of the testis-determining gene Sry in adult mouse testis. Cell 1993, 73, 1019–1030. [Google Scholar] [CrossRef]
  28. Qu, S.; Yang, X.; Li, X.; Wang, J.; Gao, Y.; Shang, R.; Sun, W. Circular RNA: A new star of non-coding RNAs. Cancer Lett. 2015, 365, 141–148. [Google Scholar] [CrossRef]
  29. Mumtaz, P.; Taban, Q.; Dar, M.; Mir, S.; Haq, Z.; Zargar, S.; Shah, R.; Ahmad, S. Deep Insights in Circular RNAs: From biogenesis to therapeutics. Biol. Proced. Online 2020, 22, 10. [Google Scholar] [CrossRef]
  30. Kos, A.; Dijkema, R.; Arnberg, A.; van der Meide, P.; Schellekens, H. The hepatitis delta (delta) virus possesses a circular RNA. Nature 1986, 323, 558–560. [Google Scholar] [CrossRef]
  31. Hansen, T.; Wiklund, E.; Bramsen, J.; Villadsen, S.; Statham, A.; Clark, S.; Kjems, J. miRNA-dependent gene silencing involving Ago2-mediated cleavage of a circular antisense RNA. EMBO J. 2011, 30, 4414–4422. [Google Scholar] [CrossRef] [PubMed]
  32. Jens, M.; Rajewsky, N. Competition between target sites of regulators shapes post-transcriptional gene regulation. Nat. Rev. Genet. 2015, 16, 113–126. [Google Scholar] [CrossRef]
  33. Bissels, U.; Wild, S.; Tomiuk, S.; Holste, A.; Hafner, M.; Tuschl, T.; Bosio, A. Absolute quantification of microRNAs by using a universal reference. RNA 2009, 15, 2375–2384. [Google Scholar] [CrossRef] [PubMed]
  34. Chao, C.; Chan, D.; Kuo, A.; Leder, P. The mouse formin (Fmn) gene: Abundant circular RNA transcripts and gene-targeted deletion analysis. Mol. Med. 1998, 4, 614–628. [Google Scholar] [CrossRef] [PubMed]
  35. Fan, X.; Zhang, X.; Wu, X.; Guo, H.; Hu, Y.; Tang, Y.; Huang, Y. Single-cell RNA-seq transcriptome analysis of linear and circular RNAs in mouse preimplantation embryos. Genome Biol. 2015, 16, 148. [Google Scholar] [CrossRef] [PubMed]
  36. Szabo, L.; Morey, R.; Palpant, N.; Wang, P.; Afari, N.; Jiang, C.; Parast, M.; Murry, C.; Laurent, L.; Salzman, J. Statistically based splicing detection reveals neural enrichment and tissue-specific induction of circular RNA during human fetal development. Genome Biol. 2015, 16, 126. [Google Scholar] [CrossRef]
  37. Zhang, Y.; Yang, H.; Long, Y.; Li, W. Circular RNA in blood corpuscles combined with plasma protein factor for early prediction of pre-eclampsia. BJOG Int. J. Obstet. Gynaecol. 2016, 123, 2113–2118. [Google Scholar] [CrossRef]
  38. Qian, Y.; Lu, Y.; Rui, C.; Qian, Y.; Cai, M.; Jia, R. Potential significance of circular RNA in human placental tissue for patients with preeclampsia. Cell Physiol. Biochem. 2016, 39, 1380–1390. [Google Scholar] [CrossRef]
  39. Qian, Y.; Wang, X.; Ruan, H.; Rui, C.; Mao, P.; Cheng, Q.; Jia, R. Circular RNAs expressed in chorionic villi are probably involved in the occurrence of recurrent spontaneous abortion. Biomed. Pharmacother. 2017, 88, 1154–1162. [Google Scholar] [CrossRef]
  40. Cheng, J.; Huang, J.; Yuan, S.; Zhou, S.; Yan, W.; Shen, W. Circular RNA expression profiling of human granulosa cells during maternal aging reveals novel transcripts associated with assisted reproductive technology outcomes. PLoS ONE 2017, 12, e0177888. [Google Scholar] [CrossRef]
  41. Zhang, S.; Ding, Y.; He, J.; Zhang, J.; Liu, X.; Chen, X.; Su, Y.; Wang, Y.; Gao, R. Altered expression patterns of circular RNAs between implantation sites and interimplantation sites in early pregnant mice. J. Cell Physiol. 2018, 234, 9862–9872. [Google Scholar] [CrossRef] [PubMed]
  42. Bai, Y.; Rao, H.; Chen, W.; Luo, X.; Tong, C.; Qi, H. Profiles of circular RNAs in human placenta and their potential roles related to preeclampsia. Biol. Reprod. 2018, 98, 705–712. [Google Scholar] [CrossRef] [PubMed]
  43. Jiang, M.; Lash, G.; Zhao, X.; Long, Y.; Guo, C.; Yang, H. CircRNA-0004904, CircRNA-0001855, and PAPP-A: Potential Novel Biomarkers for the Prediction of Preeclampsia. Cell Physiol. Biochem. 2018, 46, 2576–2586. [Google Scholar] [CrossRef] [PubMed]
  44. Zhou, W.; Wang, H.; Wu, X.; Long, W.; Zheng, F.; Kong, J.; Yu, B. The profile analysis of circular RNAs in human placenta of preeclampsia. Exp. Biol. Med. 2018, 243, 1109–1117. [Google Scholar] [CrossRef] [PubMed]
  45. Hu, X.; Ao, J.; Li, X.; Zhang, H.; Wu, J.; Cheng, W. Competing endogenous RNA expression profiling in pre-eclampsia identifies hsa_circ_0036877 as a potential novel blood biomarker for early pre-eclampsia. Clin. Epigenet. 2018, 10, 48. [Google Scholar] [CrossRef] [PubMed]
  46. Shen, X.; Zheng, L.; Huang, J.; Kong, H.; Chang, Y.; Wang, F.; Xin, H. CircTRNC18 inhibits trophoblast cell migration and epithelial–mesenchymal transition by regulating miR-762/Grhl2 pathway in pre-eclampsia. RNA Biol. 2019, 16, 1563–1573. [Google Scholar] [CrossRef]
  47. Wang, H.; She, G.; Zhou, W.; Liu, K.; Miao, J.; Yu, B. Expression profile of circular RNAs in placentas of women with gestational diabetes mellitus. Endocr. J. 2019, 22, 431–441. [Google Scholar] [CrossRef]
  48. Cao, M.; Zhang, L.; Lin, Y.; Li, Z.; Xu, J.; Shi, Z.; Chen, Z.; Ma, J.; Wen, J. Circular RNA expression profiles in umbilical cord blood exosomes from normal and gestational diabetes mellitus patients. Biosci. Rep. 2020, 40, BSR20201946. [Google Scholar] [CrossRef]
  49. Oudejans, C.; Manders, V.; Visser, A.; Keijser, R.; Min, N.; Poutsma, A.; Mulders, J.; van den Berkmortel, T.; Wigman, D.-J.; Blanken, B. Circular RNA sequencing of maternal platelets: A novel tool for the identification of pregnancy-specific biomarkers. Clin. Chem. 2021, 67, 508–517. [Google Scholar] [CrossRef]
  50. Yang, H.; Ye, W.; Chen, R.; Zeng, F.; Long, Y.Z.; Zhang, X.; Ma, J.; Gan, Q.; Rehemutula, R.; Zhu, C. Circulating expression of Hsa_circRNA_102893 contributes to early gestational diabetes mellitus detection. Sci. Rep. 2020, 10, 19046. [Google Scholar] [CrossRef]
  51. Wang, D.; Na, Q.; Song, G.; Wang, Y.; Wang, Y. The Role of circRNA-SETD2/miR-519a/PTEN Axis in Fetal Birth Weight through Regulating Trophoblast Proliferation. BioMed Res. Int. 2020, 2020, 9809632. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, H.; Zhou, W.; She, G.; Yu, B.; Sun, L. Downregulation of hsa_circ_00052.43 induces trophoblast cell dysfunction and inflammation via the β-catenin and NF-κB pathways. Reprod. Biol. Endocrinol. 2020, 18, 51. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, Y.; Yang, H.; Zhang, Y.; Shi, J.; Chen, R.; Xiao, X. CircSFXN1 regulates the behaviour of trophoblasts and likely mediates preeclampsia. Placenta 2020, 101, 115–123. [Google Scholar] [CrossRef] [PubMed]
  54. Zhou, B.; Zhang, X.; Li, T.; Xie, R.; Zhou, J.; Luo, Y.; Yang, C. CircZDHHC20 represses the proliferation, migration and invasion in trophoblast cells by miR-144/GRHL2 axis. Cancer Cell Int. 2020, 20, 19. [Google Scholar] [CrossRef] [PubMed]
  55. Zhu, H.; Niu, X.; Li, Q.; Zhao, Y.; Chen, X.; Sun, H. Circ_0085296 suppresses trophoblast cell proliferation, invasion, and migration via modulating miR-144/E-cadherin axis. Placenta 2020, 97, 18–25. [Google Scholar] [CrossRef] [PubMed]
  56. Qi, T.; Zhang, D.; Shi, X.; Li, M.; Xu, H. Decreased circUBAP2 expression is associated with preeclampsia by limiting trophoblast cell proliferation and migration. Reprod. Sci. 2021, 28, 2237–2245. [Google Scholar] [CrossRef]
  57. Li, X.; Yang, R.; Xu, Y.; Zhang, Y. Circ_0001438 participates in the pathogenesis of preeclampsia via the circ_0001438/miR-942/NLRP3 regulatory network. Placenta 2021, 104, 40–50. [Google Scholar] [CrossRef]
  58. Ma, B.; Zhao, H.; Gong, L.; Xiao, X.; Zhou, Q.; Lu, H.; Cui, Y.; Xu, H.; Wu, S.; Tang, Y.; et al. Differentially expressed circular RNAs and the competing endogenous RNA network associated with preeclampsia. Placenta 2021, 103, 232–241. [Google Scholar] [CrossRef]
  59. Tang, R.; Zhang, Z.; Han, W. CircLRRK1 targets miR-223-3p to inhibit the proliferation, migration and invasion of trophoblast cells by regulating the PI3K/AKT signaling pathway. Placenta 2021, 104, 110–118. [Google Scholar] [CrossRef]
  60. Chen, H.; Zhang, S.; Wu, Y.; Li, Z.; Wang, D.; Cai, S.; Wang, Z. The role of circular RNA circ_0008285 in gestational diabetes mellitus by regulating the biological functions of trophoblasts. Biol. Res. 2021, 54, 14. [Google Scholar] [CrossRef]
  61. Dai, W.; Liu, X. Circular RNA 0004904 promotes autophagy and regulates the fused in sarcoma/vascular endothelial growth factor axis in preeclampsia. Int. J. Mol. Med. 2021, 47, 1–10. [Google Scholar] [CrossRef]
  62. Ping, Z.; Ai, L.; Shen, H.; Zhang, X.; Jiang, H.; Song, Y. Identification and comparison of circular RNAs in preeclampsia. PeerJ 2021, 9, e11299. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, L.; Zeng, M.; Tang, F.; Chen, J.; Cao, D.; Tang, Z.-n. Circ-PNPT1 contributes to gestational diabetes mellitus (GDM) by regulating the function of trophoblast cells through miR-889-3p/PAK1 axis. Diabetol. Metab. Syndr. 2021, 13, 1–14. [Google Scholar] [CrossRef]
  64. Tang, M.; Bai, L.; Wan, Z.; Wan, S.; Xiang, Y.; Qian, Y.; Cui, L.; You, J.; Hu, X.; Qu, F. circRNA-DURSA regulates trophoblast apoptosis via miR-760-HIST1H2BE axis in unexplained recurrent spontaneous abortion. Mol. Ther. Nucleic Acids 2021, 26, 1433–1445. [Google Scholar] [CrossRef]
  65. Yao, P.; Hu, G.; Niu, H. Hsa_circ_0074371 Regulates Proliferation, Apoptosis, Migration, and Invasion via the miR-582-3p/LRP6 Axis in Trophoblast Cells. Biochem. Genet. 2021, 60, 267–285. [Google Scholar] [CrossRef] [PubMed]
  66. Fan, Z.; Wang, Q.; Deng, H. Circ_0011460 upregulates HTRA1 expression by sponging miR-762 to suppress HTR8/SVneo cell growth, migration, and invasion. Am. J. Reprod. Immunol. 2021, 86, e13485. [Google Scholar] [CrossRef] [PubMed]
  67. Li, C.; Li, Q. Circular RNA circ_0111277 serves as ceRNA, targeting the miR-424-5p/NFAT5 axis to regulate the proliferation, migration, and invasion of trophoblast cells in preeclampsia. Reprod. Sci. 2021, 29, 923–935. [Google Scholar] [CrossRef]
  68. Zhou, F.; Liu, H.; Zhang, R.; Sun, Y. Circ_0007121 Facilitates Trophoblastic Cell Proliferation, Migration, and Invasion via the Regulation of the miR-421/ZEB1 Axis in Preeclampsia. Reprod. Sci. 2022, 29, 100–109. [Google Scholar] [CrossRef]
  69. Huang, Y.; Liang, B.; Chen, X. Exosomal circular RNA circ_0074673 regulates the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells via the microRNA-1200/MEOX2 axis. Bioengineered 2021, 12, 6782–6792. [Google Scholar] [CrossRef]
  70. Shan, L.; Hou, X. Circular RNA hsa_circ_0026552 inhibits the proliferation, migration and invasion of trophoblast cells via the miR-331-3p/TGF-βR1 axis in pre-eclampsia. Mol. Med. Rep. 2021, 24, 798. [Google Scholar] [CrossRef]
  71. Jiang, B.; Zhang, J.; Sun, X.; Yang, C.; Cheng, G.; Xu, M.; Li, S.; Wang, L. Circulating exosomal hsa_circRNA_0039480 is highly expressed in gestational diabetes mellitus and may be served as a biomarker for early diagnosis of GDM. J. Transl. Med. 2022, 20, 5. [Google Scholar] [CrossRef] [PubMed]
  72. Gao, Y.; Tang, Y.; Sun, Q.; Guan, G.; Wu, X.; Shi, F.; Zhou, Z.; Yang, W. Circular RNA FOXP1 relieves trophoblastic cell dysfunction in recurrent pregnancy loss via the miR-143-3p/S100A11 cascade. Bioengineered 2021, 12, 9081–9093. [Google Scholar] [CrossRef] [PubMed]
  73. Wu, H.; Zheng, X.; Liu, Y.; Shen, J.; Ye, M.; Zhang, Y. Hsa_circRNA_102682 is closely related to lipid metabolism in gestational diabetes mellitus. J. Gynaecol. Endocrinol. 2022, 38, 50–54. [Google Scholar] [CrossRef] [PubMed]
  74. She, W.; Li, T.; Liu, Y.; Liu, X. CircRNA circVEGFC is Highly Expressed in Gestational Diabetes Mellitus (GDM) and It is Correlated with Multiple Adverse Events. Diabetes Metab. Syndr. Obes. 2021, 14, 4409. [Google Scholar] [CrossRef] [PubMed]
  75. Zou, H.; Mao, Q. Circ_0037078 promotes trophoblast cell proliferation, migration, invasion and angiogenesis by miR-576-5p/IL1RAP axis. Am. J. Reprod. Immunol. 2022, 87, e13507. [Google Scholar] [CrossRef]
  76. Mao, Q.; Zou, H. Circular RNA circ_0032962 promotes trophoblast cell progression as ceRNA to target PBX3 via sponging miR-326 in preeclampsia. Reprod. Biol. 2021, 21, 100571. [Google Scholar] [CrossRef]
  77. Wang, L.; Wang, X.; Chen, X.; Wu, D.; Cen, H.; Mao, D.; Mo, Y.; Zheng, L. The relationship between hsa_circ_0051326 and HLA-G expression in the blood of patients with pre-eclampsia. Ginekol. Pol. 2021, in press. [Google Scholar] [CrossRef]
  78. Shu, C.; Xu, P.; Han, J.; Han, S.; He, J. Upregulation of circRNA hsa_circ_0008726 in Pre-eclampsia Inhibits Trophoblast Migration, Invasion, and EMT by Regulating miR-345-3p/RYBP Axis. Reprod. Sci. 2021, in press. [Google Scholar] [CrossRef]
  79. Zhang, Y.; Yang, H.; Long, Y.; Zhang, Y.; Chen, R.; Shi, J.; Chen, J. circRNA N6-methyladenosine methylation in preeclampsia and the potential role of N6-methyladenosine-modified circPAPPA2 in trophoblast invasion. Sci. Rep. 2021, 11, 24357. [Google Scholar] [CrossRef]
  80. Wang, W.; Liu, J.; Pan, E. CircHIPK3 contributes to human villous trophoblast growth, migration and invasion via modulating the pathway of miR-346/KCMF1. Placenta 2021, 118, 46–54. [Google Scholar] [CrossRef]
  81. Li, J.; Han, J.; Zhao, A.; Zhang, G. CircPAPPA Regulates the Proliferation, Migration, Invasion, Apoptosis, and Cell Cycle of Trophoblast Cells Through the miR-3127-5p/HOXA7 Axis. Reprod. Sci. 2022, 29, 1215–1225. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, W.; Shi, J.; Zheng, L. Identification of Circular RNA circ_0017068 as a Regulator of Proliferation and Apoptosis in Trophoblast Cells by miR-330-5p/XIAP Axis. Reprod. Sci. 2022, in press. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, Y.; Yang, H.; Zhang, Y.; Shi, J.; Long, Y. A Novel Circular RNA CircBRAP May Be Used as an Early Predictor of Preeclampsia and Its Potential Mechanism. Reprod. Sci. 2022, in press. [Google Scholar] [CrossRef] [PubMed]
  84. Yuan, Y.; Gong, Y.; Zhong, L.; Ding, X.; Yang, Z.; Su, X.; Chen, M.; Zhang, F.; Yang, L. Circular RNA expression profile and competing endogenous RNA regulatory network in preeclampsia. Placenta 2022, 19, 32–38. [Google Scholar] [CrossRef]
  85. Wang, H.; Zhang, Y.; Zhang, J.; Du, X.; Li, Q.; Pan, Z. circSLC41A1 Resists Porcine Granulosa Cell Apoptosis and Follicular Atresia by Promoting SRSF1 through miR-9820-5p Sponging. Int. J. Mol. Sci. 2022, 23, 1509. [Google Scholar] [CrossRef]
  86. Pandey, P.; Rout, P.; Das, A.; Gorospe, M.; Panda, A. RPAD (RNase R treatment, polyadenylation, and poly(A)+ RNA depletion) method to isolate highly pure circular RNA. Methods 2019, 155, 41–48. [Google Scholar] [CrossRef]
  87. Xiao, M.; Wilusz, J. An improved method for circular RNA purification using RNase R that efficiently removes linear RNAs containing G-quadruplexes or structured 3’ ends. Nucleic Acids Res. 2019, 41, 8755–8769. [Google Scholar] [CrossRef]
  88. Holtan, S.; Creedon, D.; Haluska, P.; Markovic, S. Cancer and pregnancy: Parallels in growth, invasion, and immune modulation and implications for cancer therapeutic agents. Mayo Clin. Proc. 2009, 84, 985–1000. [Google Scholar] [CrossRef]
  89. Costanzo, V.; Bardelli, A.; Siena, S.; Abrignani, S. Exploring the links between cancer and placenta development. Open Biol. 2018, 8, 180081. [Google Scholar] [CrossRef]
  90. Bischof, P.; Campana, A. A putative role for oncogenes in trophoblast invasion? Mol. Hum. Reprod. 2000, 15, 51–58. [Google Scholar]
  91. Kim, D.; Xing, T.; Yang, Z.; Dudek, R.; Lu, Q.; Chen, Y. Epithelial mesenchymal transition in embryonic development, tissue repair and cancer: A comprehensive overview. J. Clin. Med. 2017, 7, 1. [Google Scholar] [CrossRef] [PubMed]
  92. Hanahan, D.; Weinberg, R. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  93. Hammer, A. Immunological regulation of trophoblast invasion. J. Reprod. Immunol. 2011, 90, 21–28. [Google Scholar] [CrossRef] [PubMed]
  94. Demir, R.; Kaufmann, P.; Castellucci, M.; Erbengi, T.; Kotowski, A. Fetal vasculogenesis and angiogenesis in human placental villi. Acta Anat. (Basel) 1989, 136, 190–203. [Google Scholar] [CrossRef] [PubMed]
  95. Morrish, D.; Bhardwaj, D.; Dabbagh, L.; Marusyk, H.; Siy, O. Epidermal growth factor induces differentiation and secretion of human chorionic gonadotropin and placental lactogen in normal human placenta. J. Clin. Endocrinol. Metab. 1987, 65, 1282–1290. [Google Scholar] [CrossRef]
  96. Normanno, N.; Bianco, C.; De Luca, A.; Salomon, D. The role of EGF-related peptides in tumor growth. Front. Biosci. 2001, 6, 685–707. [Google Scholar] [CrossRef]
  97. Xi, J.; Wang, Y.; Long, X.; Ma, Y. Mangiferin potentiates neuroprotection by isoflurane in neonatal hypoxic brain injury by reducing oxidative stress and activation of phosphatidylinositol-3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling. Med. Sci. Monit. 2018, 24, 7459–7468. [Google Scholar] [CrossRef]
  98. Nicholson, K.; Anderson, N. The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal. 2002, 14, 381–395. [Google Scholar] [CrossRef]
  99. Marzusch, K.; Ruckh, P.; Horny, H.; Dietle, J.; Kaiserling, E. Expression of the p53 Tumour Suppressor Gene in Human Placenta: An Immunohistochemical Study. Placenta 1995, 16, 101–104. [Google Scholar] [CrossRef]
  100. Lane, D. Mutation of the p53 protein: Common steps found in the majority of human cancers. In Accomplishments in Cancer Research; Fortner, J.G., Ed.; Lippincott Co: Philadelphia, PA, USA, 1990; pp. 252–266. [Google Scholar]
  101. Forbes, K.; Westwood, M.; Baker, P.N.; Aplin, J.D. Insulin-like growth factor I and II regulate the life cycle of trophoblast in the developing human placenta. Am. J. Physiol. Cell Physiol. 2008, 294, 1313–1322. [Google Scholar] [CrossRef]
  102. Jones, J.; Clemmons, D. Insulin-like growth factors and their binding proteins: Biological actions. Endocr. Rev. 1995, 16, 3–35. [Google Scholar] [PubMed]
  103. Dufourny, B.; Alblas, J.; van Teeffelen, H.; van Schaik, F.; van der Burg, B.; Steenbergh, P. Mitogenic signaling of insulin-like growth factor I in MCF-7 human breast cancer cells requires phosphatidylinositol 3-kinase and is independent of mitogen-activated protein kinase. J. Biol. Chem. 1997, 272, 31163–31171. [Google Scholar] [CrossRef] [PubMed]
  104. Buckbinder, L.; Talbott, R.; Velasco Miguel, S.; Takenaka, I.; Faha, B.; Seizinger, B. Induction of the growth inhibitor IGF-binding protein 3 by p53. Nature 1995, 377, 646–649. [Google Scholar] [CrossRef] [PubMed]
  105. Gorivodsky, M.; Torchinsky, A.; Zemliak, I.; Savion, S.; Fein, A.; Toder, V. TGF beta 2 mRNA expression and pregnancy failure in mice. Am. J. Reprod. Immunol. 1999, 42, 124–133. [Google Scholar] [PubMed]
  106. Akhurst, R.; Derynck, R. TGF-β signaling in cancer—A double-edged sword. Trends Cell Biol. 2001, 11, 44–51. [Google Scholar]
  107. Ahmed, A.; Dunk, C.; Kniss, D.; Wilkes, M. Role of VEGF receptor (Flt-1) in mediating calcium dependant nitric oxide release and limiting DNA synthesis in human trophoblast cells. Lab. Invest. 1997, 76, 779–791. [Google Scholar]
  108. Senger, D.; Galli, S.; Dvorak, A.; Perruzzi, C.; Harvey, V.; Dvorak, H. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science 1983, 219, 983–985. [Google Scholar] [CrossRef]
  109. Gangloff, Y.; Mueller, M.; Dann, S.; Svoboda, P.; Sticker, M.; Spetz, J.; Um, S.; Brown, E.; Cereghini, S.; Thomas, G.; et al. Disruption of the mouse mTOR gene leads to early postimplantation lethality and prohibits embryonic stem cell development. Mol. Cell 2004, 24, 9508–9516. [Google Scholar] [CrossRef]
  110. Martin, P.; Sutherland, A. Exogenous amino acids regulate trophectoderm differentiation in the mouse blastocyst through an mTOR-dependent pathway. Dev. Biol. 2001, 240, 182–193. [Google Scholar] [CrossRef]
  111. Faivre, S.; Kroemer, G.; Raymond, E. Current development of mTOR inhibitors as anticancer agents. Nat. Rev. Drug Discov. 2006, 5, 671–688. [Google Scholar] [CrossRef]
  112. Nadeau, V.; Guillemette, S.; Belanger, L.; Jacob, O.; Roy, S.; Charron, J. Map2k1 and Map2k2 genes contribute to the normal development of syncytiotrophoblasts during placentation. Development 2009, 136, 1363–1374. [Google Scholar] [CrossRef]
  113. Plotnikov, A.; Zehorai, E.; Procaccia, S.; Seger, R. The MAPK cascades: Signaling components, nuclear roles and mechanisms of nuclear translocation. Biochim. Biophys. Acta 2011, 1813, 1619–1633. [Google Scholar] [CrossRef] [PubMed]
  114. Fitzgerald, J.; Germeyer, A.; Huppertz, B.; Jeschke, U.; Knofler, M.; Moser, G. Governing the invasive trophoblast: Current aspects on intra- and extra-cellular regulation. Am. J. Reprod. Immunol. 2010, 63, 492–505. [Google Scholar] [CrossRef] [PubMed]
  115. Thomas, S.; Snowden, J.; Zeidler, M.; Danson, S. The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br. J. Cancer 2015, 113, 365–371. [Google Scholar] [CrossRef] [PubMed]
  116. Vainio, S.; Heikkila, M.; Kispert, A.; Chin, N.; McMahon, A. Female development in mammals is regulated by Wnt-4 signalling. Nature 1999, 397, 405–409. [Google Scholar] [CrossRef]
  117. Nusse, R.; van Ooyen, A.; Cox, D.; Fung, Y.; Varmus, H. Mode of proviral activation of a putative mammary oncogene (int-1) on mouse chromosome 15. Nature 1984, 307, 131–136. [Google Scholar] [CrossRef]
  118. Lindstrom, T.; Bennett, P. The role of nuclear factor kappa B in human labour. Reproduction 2005, 130, 569–581. [Google Scholar] [CrossRef]
  119. Pikarsky, E.; Porat, R.; Stein, I.; Abramovitch, R.; Amit, S.; Kasem, S.; Gutkovich-Pyest, E.; Urieli-Shoval, S.; Galun, E.; Ben-Neriah, Y. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004, 431, 461–466. [Google Scholar] [CrossRef]
  120. Pique-Regi, R.; Romero, R.; Tarca, A.L.; Sendler, E.D.; Xu, Y.; Garcia-Flores, V.; Leng, Y.; Luca, F.; Hassan, S.S.; Gomez-Lopez, N. Single cell transcriptional signatures of the human placenta in term and preterm parturition. Elife 2019, 8, e52004. [Google Scholar] [CrossRef]
  121. Suryawanshi, H.; Morozov, P.; Straus, A.; Sahasrabudhe, N.; Max, K.E.; Garzia, A.; Kustagi, M.; Tuschl, T.; Williams, Z. A single-cell survey of the human first-trimester placenta and decidua. Sci. Adv. 2018, 4, eaau4788. [Google Scholar] [CrossRef]
  122. Gong, S.; Gaccioli, F.; Dopierala, J.; Sovio, U.; Cook, E.; Volders, P.-J.; Martens, L.; Kirk, P.D.; Richardson, S.; Smith, G. The RNA landscape of the human placenta in health and disease. Nat. Commun. 2021, 12, 2639. [Google Scholar] [CrossRef] [PubMed]
  123. Buckberry, S.; Bianco-Miotto, T.; Bent, S.J.; Clifton, V.; Shoubridge, C.; Shankar, K.; Roberts, C.T. Placental transcriptome co-expression analysis reveals conserved regulatory programs across gestation. BMC Genet. 2017, 18, 10. [Google Scholar] [CrossRef] [PubMed]
  124. Mikheev, A.; Nabekura, T.; Kaddoumi, A.; Bammler, T. Profiling gene expression in human placentae of different gestational ages: An OPRU Network and UW SCOR Study. Reprod. Sci. 2008, 15, 866–877. [Google Scholar] [CrossRef] [PubMed]
  125. Zhong, Z.; Huang, M.; Lv, M.; He, Y.; Duan, C.; Zhang, L. Circular RNA MYLK as a competing endogenous RNA promotes bladder cancer progression through modulating VEGFA/VEGFR2 signaling pathway. Placenta 2017, 403, 305–317. [Google Scholar] [CrossRef]
  126. Zeman, M.; Cimprich, K. Causes and consequences of replication stress. Nat. Cell Biol. 2014, 16, 2–9. [Google Scholar] [CrossRef]
  127. Conn, V.M.; Hugouvieux, V.; Nayak, A.; Conos, S.A.; Capovilla, G.; Cildir, G.; Jourdain, A.; Tergaonkar, V.; Schmid, M.; Zubieta, C.; et al. A circRNA from SEPALLATA3 regulates splicing of its cognate mRNA through R-loop formation. Nat. Plants 2017, 3, 17053. [Google Scholar] [CrossRef]
  128. Weigelt, C.; Sehgal, R.; Tain, L.; Cheng, J.; Eßer, J.; Pahl, A.; Dieterich, C.; Gronke, S.; Partridge, L. An Insulin-Sensitive Circular RNA that Regulates Lifespan in Drosophila. Mol. Cell. 2020, 79, 268–279. [Google Scholar] [CrossRef]
  129. Knupp, D.; Miura, P. CircRNA accumulation: A new hallmark of aging? Mech. Ageing Dev. 2018, 173, 71–79. [Google Scholar] [CrossRef]
  130. Maiti, K.; Sultana, Z.; Aitken, R.J.; Morris, J.; Park, F.; Andrew, B.; Riley, S.; Smith, R. Evidence that fetal death is associated with placental aging. Am. J. Obstet. Gynecol. 2017, 217, 441.e1–441.e14. [Google Scholar] [CrossRef]
Figure 1. An example of circRNA biogenesis using backsplicing. circRNAs are produced through backsplicing and can potentially produce several alternatively spliced products. circRNAs can: (A) comprise multiple exons, (B,C) comprise a single exon, (D) comprise both exons and introns (termed exonic intronic circRNAs) or (E) comprise only introns (termed circular intronic RNAs). After each backsplicing event, the remaining exons are left to form an alternatively spliced transcript. Graphic created with BioRender.com, accessed on 5 May 2021.
Figure 1. An example of circRNA biogenesis using backsplicing. circRNAs are produced through backsplicing and can potentially produce several alternatively spliced products. circRNAs can: (A) comprise multiple exons, (B,C) comprise a single exon, (D) comprise both exons and introns (termed exonic intronic circRNAs) or (E) comprise only introns (termed circular intronic RNAs). After each backsplicing event, the remaining exons are left to form an alternatively spliced transcript. Graphic created with BioRender.com, accessed on 5 May 2021.
Ijms 23 04551 g001
Figure 2. Methods of circRNA biogenesis. A number of different methods facilitate the circularisation of circRNAs: (A) Complementary base-pairing (e.g., via Alu repeats) promotes backsplicing due to spatial reduction in the splice sites. (B) RBP-driven circularisation occurs when RBPs bind flanking introns and bridge them together for splicing. (C) ciRNA formation: ciRNAs are formed from lariat introns that escape debranching. C-rich (red) and GU-rich (blue) sequence binding is sufficient for the intron to avoid debranching and generate a ciRNA. (D) The lariat-driven model of circularisation. Exon-skipping occurs to bring splice sites into close proximity.
Figure 2. Methods of circRNA biogenesis. A number of different methods facilitate the circularisation of circRNAs: (A) Complementary base-pairing (e.g., via Alu repeats) promotes backsplicing due to spatial reduction in the splice sites. (B) RBP-driven circularisation occurs when RBPs bind flanking introns and bridge them together for splicing. (C) ciRNA formation: ciRNAs are formed from lariat introns that escape debranching. C-rich (red) and GU-rich (blue) sequence binding is sufficient for the intron to avoid debranching and generate a ciRNA. (D) The lariat-driven model of circularisation. Exon-skipping occurs to bring splice sites into close proximity.
Ijms 23 04551 g002
Figure 3. circRNA functions. circRNAs are able to complete numerous roles: (A) Translation can occur in the presence of an IRES. (B) circRNAs can function as miRNA sponges, by “mopping up” miRNAs and preventing their actions. (C) mRNA traps (inhibits translation) by sequestering the translation start site on mRNA. (D) circRNAs are also able to bind proteins, including RBPs and (E) enzyme–substrate complexes, to facilitate actions (denoted with **) such as phosphorylation, ubiquitylation and acetylation. (F) circRNAs can form circ-R-loops with DNA and impede transcription, facilitating DNA breaks. (G) circRNAs can also influence the host promoter region, altering DNA replication and transcription.
Figure 3. circRNA functions. circRNAs are able to complete numerous roles: (A) Translation can occur in the presence of an IRES. (B) circRNAs can function as miRNA sponges, by “mopping up” miRNAs and preventing their actions. (C) mRNA traps (inhibits translation) by sequestering the translation start site on mRNA. (D) circRNAs are also able to bind proteins, including RBPs and (E) enzyme–substrate complexes, to facilitate actions (denoted with **) such as phosphorylation, ubiquitylation and acetylation. (F) circRNAs can form circ-R-loops with DNA and impede transcription, facilitating DNA breaks. (G) circRNAs can also influence the host promoter region, altering DNA replication and transcription.
Ijms 23 04551 g003
Table 1. Summary of research on circRNAs in female reproductive tissues and blood during pregnancy (limited to studies using primary tissue).
Table 1. Summary of research on circRNAs in female reproductive tissues and blood during pregnancy (limited to studies using primary tissue).
AuthorYearTissuePregnancy StatusKey FindingsLimitations
Fan X, et al. [35]2015Mouse—oocytes and preimplantation embryosUncomplicated (assumed)Detected 2891 circRNAs from 1316 host genes. A majority of these circRNAs are unique to the preimplantation stage and a large proportion of them exhibit dynamic expression patterns during this developmental process.Sequencing using the SUPeR-seq method could possibly limit depth of circRNA sequencing due to no enrichment using RNase R.
Szabo L, et al. [36]2015Fetal tissue—unspecified, various speciesUncomplicated (assumed)Developed an algorithm to compare established data sets in human, rat and mouse tissue and cell lines, with their generated circRNA data from RNA-seq on fetal tissue.This study assumes a degree of conservation between species.
Zhang YG, et al. [37]2016Human maternal red blood cellsPE vs. uncomplicatedThe levels of circ_101222 in red blood cells of patients with PE were significantly higher than in healthy women. Using ENG in combination with circ_101222 improved confidence for the prediction of PE.
Qian Y, et al. [38]2016Human placentaPE vs. preterm birth (PTB)143 circRNAs were up-regulated and 158 were down-regulated in PE samples compared with preterm.Use of microarray is not as comprehensive as sequencing techniques. PTB placenta is used as a gestational age control but PTB can occur for multiple reasons and is a pathology of pregnancy. Caution on interpretation is required.
Qian Y, et al. [39]2017Human placenta—chorionic villiRecurrent spontaneous abortion (RSA) vs uncomplicated594 aberrantly expressed circRNAs between gestational age-matched RSA and healthy placentae. Of these, 335 circRNAs were up-regulated and 259 were down-regulated.No validation or investigation into results or mechanisms.
Cheng J, et al. [40]2017Human granulosa cellsNon-pregnant
Advanced age (AA ≥ 38 years) vs. young age (YA ≤ 30 years)
46 upregulated and 11 downregulated circRNAs in AA samples compared with YA.Use of microarray is not as thorough as sequencing techniques.
Zhang S, et al. [41]2018Mouse—endometriumUncomplicated (assumed)Used microarray to find that 101 upregulated and 75 downregulated circRNAs at implantation sites compared with interimplantation sites. Four randomly selected circRNAs were validated for their expression using qRT-PCRUse of microarray is not as comprehensive as sequencing techniques.
Bai Y, et al. [42]2018Human placentaPE vs. uncomplicated151 circRNAs were upregulated and 149 were downregulated in PE samples compared with normal. Possible biomarker hsa_circ_0007121 had a significant predictive indexOnly 3 of the 10 circRNAs for validation matched sequencing results.
Jiang M, et al. [43]2018Human maternal peripheral blood mononuclear cellsPE vs. uncomplicated884 circRNAs were downregulated and 1294 circRNAs were upregulated in PE samples compared with control. circ_0004904 and circ_0001855 combined with PAPP-A might be biomarkers for PE detection.Use of microarray is not as thorough as sequencing techniques. As blood was collected at the time of disease, there is no way to be certain that circ_0004904 and circ_0001855 are causes of PE and not resultant.
Zhou W, et al. [44]2018Human placentaPE vs. uncomplicatedTwo circRNAs were up-regulated and 47 were down-regulated in PE compared with control placentae. Hsa_circRNA_3286 reduced invasion in HTR-8/SVneo cells.RNase R enrichment for circRNAs not completed on RNA-seq samples. Only 3 of the 10 circRNAs for validation matched sequencing results.
Hu X, et al. [45]2018Human placenta
Maternal whole blood
Severe PE vs. uncomplicated4569 upregulated and 3984 downregulated circRNAs between severe PE and healthy pregnancy. Identified hsa_circ_0036877 as a potential novel blood biomarker for early PE.Use of microarray is not as thorough as sequencing techniques. Significant differences in BMI, gestational age at delivery and % caesarean sections between severe PE and control groups.
Shen, XY, et al. [46]2019Human placentaPE vs. uncomplicatedcircTNRC18 upregulated in PE placentae. circTNRC18 reduced trophoblast cell migration and EMT. circTNRC18 repressed miR-762 activity and elevated Grhl2 protein.
Wang H, et al. [47]2019Human placentaGDM vs. uncomplicatedThree circRNAs were upregulated and 43 were downregulated between GDM and normal placentae. Ten randomly selected circRNAs were validated for their expression using qRT-PCR.RNase R enrichment for circRNAs not completed on RNA-seq samples. Only 3 of the 10 circRNAs for validation matched sequencing results.
Cao M, et al. [48]2020Human umbilical cord bloodGDM vs. uncomplicated229 upregulated and 278 downregulated circRNAs between GDM and healthy pregnancy. Exosome particle size was larger and exosome concentration was higher in GDM.Use of microarray is not as thorough as sequencing techniques. RNase R enrichment for circRNAs not completed on microarray samples.
Oudejans C, et al. [49]2020Human maternal plateletsUncomplicated (assumed)Proof of concept study showing that pregnancy-specific circRNAs can be detected in first-trimester platelet RNA.Could possibly limit depth of circRNA sequencing due to no enrichment using RNase R.
Yang H, et al. [50]2020Human maternal bloodGDM vs uncomplicatedBlood samples (n = 12) were collected from GDM and healthy pregnant women between 15–24 weeks’ gestation prior to RNase R treatment and circRNA microarray analysis.Use of microarray for detection of circRNAs is not as thorough as sequencing techniques. No mention of gestational age matching between GDM and controls.
Wang D, et al. [51]2020Human placentaMacrosomia vs. uncomplicatedCirc-SETD2 was upregulated in placentae of patients with fetal macrosomia. Circ-SETD2 upregulation increased proliferation and invasion in HTR-8/SVneo cells. Suggested circ-SETD2/miR-519a/PTEN axis involved in regulating trophoblasts in macrosomia.Use of lnc-microarray for detection of circRNAs is not as thorough as sequencing techniques.
Wang H, et al. [52]2020Human placenta and maternal plasmaGDM vs. uncomplicatedCirc_0005243 was identified using RNase R (determining circular form). Circ_0005243 expression was downregulated in placenta and maternal plasma in GDM. Knockdown of circ_0005243 in HTR-8/SVneo cells suppressed cell proliferation and migration and increased secretion of inflammatory factors (TNF-α and IL-6). It also reduced β-catenin expression and increased nuclear NF-κB p65 nuclear translocation.
Zhang Y, et al. [53]2020Human placenta and in vivo rat modelPE vs. uncomplicatedCircSFXN1 was identified using RNase R (determining circular form). CircSFXN1 was elevated in PE placenta. Knockdown of circSFXN1 promoted TEV-1 cell invasion and HUVEC angiogenesis—this effect was opposed with circSFXN1 overexpression. Pregnant rats injected with sFLT1-expressing adenovirus had in increased blood pressure and proteinuria; si-circSFXN1 reversed this. CircSFXN1 recruits sFLT1, validated by RNA-protein pulldown, RNA immunoprecipitation and dual-luciferase reporter assays.Use of microarray is not as thorough as sequencing techniques, although the study validates these results with qRT-PCR.
Zhou B, et al. [54]2020Human placentaPE vs. uncomplicatedCircZDHHC20 was identified using RNase R (determining circular form). CircZDHHC20 was up-regulated and miR-144 was down-regulated in PE placenta. CircZDHHC20 overexpression in HTR-8/SVneo cells repressed trophoblast proliferation, migration, and invasion. miR-144 regulated circZDHHC20 was inhibited by GRHL2.
Zhu H, et al. [55]2020Human placentaPE vs. uncomplicatedCirc_0085296 was identified using RNase R (determining circular form). Circ_0085296 elevated in PE placenta. Knockdown of circ_0085296 in HTR-8/SVneo cells promoted trophoblast cell proliferation, invasion, and migration. miR-144 down-regulated in PE placenta, directly bound to circ_0085296 and E-cadherin. Circ_0085296 bound to miR-144 to regulate E-cadherin.
Qi T, et al. [56]2020Human placentaPE vs. uncomplicatedCircUBAP2 (hsa_circ_0003496) was downregulated in PE placentae. CircUBAP2 knockdown suppressed HTR-8/SVneo cell proliferation and migration. CircUBAP2 sponges miR-1244 to regulate FOXM1. Cotransfection of si-circUBAP2 and a miR-1244 inhibitor partially restored cell proliferation and migration induced by circUBAP2 depletion.
Li X, et al. [57]2021Human placentaPE vs. uncomplicatedCirc_0001438 and NLRP3 were elevated in PE placenta. Knockdown of circ_0001438 promoted cell proliferation, migration and invasion but inhibited apoptosis and inflammatory responses in
HTR-8/SVneo cells. Circ_0001438 bound to miR-942 to regulate NLRP3.
RNase R treatment not used—Circ_0001438 was not enriched for circular form only.
Ma B, et al. [58]2021Human placentaPE vs. uncomplicated252 upregulated and 109 downregulated circRNAs between preeclamptic and healthy placentae; 6 circRNAs were further validated using qPCR.No mention of gestational age matching between PE and controls.
Tang R, et al. [59]2021Human placentaPE vs. uncomplicatedCircLRRK1 was identified using Actinomycin D and RNase R (determining circular form). circLRRK1 was elevated in PE placenta. Knockdown of circLRRK1 promoted cell proliferation, migration and invasion in HTR-8/SVneo cells. circLRRK1 bound to miR-223-3p to regulate PI3K/Akt signalling.
Chen H, et al. [60]2021Maternal plasmaGDM vs. uncomplicatedCirc_0008285 was increased, while circ_0001173 was decreased, in GDM samples. Circ_0008285 correlated with total cholesterol and LDL-C levels. Circ_0001173 correlated with glycated haemoglobin. High glucose media promoted HTR-8/SVneo cell proliferation, invasion, and migration, while circ_0008285 knockdown exerted the opposite effect.High glucose media contained 30 mmol/L glucose which is potentially too high for physiological relevance.
Dai W, et al. [61]2021Human placenta and maternal plasmaPE vs. uncomplicatedCirc_0004904 levels were elevated in PE placentae and maternal plasma. Aberrant circ_0004904 expression inhibited autophagy and induced JEG3 cell proliferation and invasion. Circ_0004904 also regulated ATG12 levels via miR-570, as well as controlling the FUS/VEGF axis in HTR-8/SVneo and JEG3 cells.
Ping Z, et al. [62]2021Maternal bloodPE vs. uncomplicated121 differentially expressed circRNAs were upregulated and 30 downregulated in PE samples. Functional and pathway enrichment analysis was conducted using Gene Ontology and KEGG databases.Could possibly limit depth of circRNA sequencing due to no enrichment using RNase R. No mechanistic studies conducted or PCR validation of results.
Zhang L, et al. [63]2021Human placentaGDM vs. uncomplicatedCirc-PNPT1 levels were elevated in GDM placentae and high glucose (HG)-induced HTR-8/SVneo cells. HG-induced arrest of cell viability, migration, invasion and apoptosis was reversed with circ-PNPT1 knockdown. Circ-PNPT1 also sponged miR-889-3p to regulate PAK1. HTR-8/SVneo cell line experiments showed circ-PNPT1 was packaged into exosomes and internalised by surrounding cells.Exosome studies were conducted in HTR-8/SVneo cell line, not replicated in placental explants or maternal blood. High glucose media contained 25 mmol/L glucose which is potentially too high for physiological relevance.
Tang M, et al. [64]2021Human placental villous tissue and embryos
In vivo mouse model
Unexplained recurrent spontaneous abortion (URSA) vs. uncomplicatedCirc-0050703 (circRNA-DURSA) is downregulated in URSA placental villous tissue. In vitro, circRNA-DURSA silencing results in cell apoptosis and circRNA-DURSA competitively binds miR-760, regulating HIST1H2BE. In vivo, circRNA-DURSA silencing decreased number of embryos successfully implanted.
Yao P, et al. [65]2021Human placentaFetal growth restriction vs. uncomplicatedCirc_0074371 and LRP6 were downregulated, and miR-582-3p was upregulated in fetal growth restriction (FGR) placentae and HTR-8/SVneo cells. Circ_0074371 sponges miR-582-3p to regulate LRP6. Circ_0074371 knockdown induced HTR-8/SVneo cell cycle arrest, apoptosis, and inhibited cell proliferation, migration, and invasion, which was reversed with a miR-582-3p inhibitor.
Fan Z, et al. [66]2021Human placentaPE vs. uncomplicatedCirc_0011460 is upregulated in PE placentae, and overexpression in HTR-8/SVneo cells suppressed proliferation, migration and invasion, and increased cell apoptosis. Circ_0011460 also sponges miR-762 and regulates HTRA1.
Li C, et al. [67]2021Human placentaPE vs. uncomplicatedCirc_0111277 and NFAT5 expression were increased in PE placentae and miR-424-5p was decreased. Circ_0111277
knockdown increased cell viability, migration, invasion, and angiogenesis in HTR-8/SVneo cells. Circ_0111277 acted as a sponge of miR-424-5p to regulate NFAT5 expression.
Zhou F, et al. [68]2022Human placentaPE vs. uncomplicated Circ_0007121 is downregulated in PE placentae. Upregulation of circ_0007121 promotes cell proliferation, migration, invasion and EMT. Circ_0007121 also sponges miR-421 and thus regulates ZEB1 expression.
Huang Y, et al. [69]2021Human umbilical cord blood—exosomes isolatedGDM vs. uncomplicatedLarger exosomes and greater number of exosomes in umbilical cord blood of GDM patients. Circ_0074673 was upregulated in exosomes from GDM and in HUVECs co-cultured with exosomes. Loss of exosomal circ_0074673 facilitated the proliferation, migration, and angiogenesis of high glucose-HUVECs via the miR- 1200/MEOX2 axis.High glucose media contained 25 mM glucose which is potentially too high for physiological relevance.
Shan L, et al. [70]2021Human placentaPE vs. uncomplicatedmiR-331-3p negatively correlates with circ_0026552 relative expression, while TGF-βR1 positively correlates with circ_0026552 expression. Silencing circ_0026552 increased proliferation, migration and invasion of HTR-8/SVneo cells, which was reversed with circ_0026552 overexpression. Circ_0026552 sponges miR-331-3p to upregulate TGF-βR1 expression.RNase R enrichment for circRNAs not completed on microarray samples. Use of microarray is not as thorough as sequencing techniques, although the study validates these results with qRT-PCR. Small number (n = 3 PE, n = 4 control) tissues used for microarray.
Jiang B, et al. [71]2021Maternal bloodGDM vs. uncomplicatedPlasma exosomal circRNA_0039480 and circRNA_0026497 were increased in GDM. circRNA_0039480 was elevated in GDM vs. normal glucose tolerance control throughout trimesters and positively correlated with OGTT during the second trimester. The combination of circRNA_0039480 and circRNA_0026497 suggested as a useful biomarker for GDM in the first trimester
(AUC = 0.754, p < 0.001).
RNase R enrichment for circRNAs not completed on microarray samples. Use of microarray is not as thorough as sequencing techniques, although the study validates these results with qRT-PCR. Small number (n = 3) samples used for microarray.
Gao Y, et al. [72]2021Human placentaRecurrent pregnancy loss (RPL) vs. uncomplicatedMiR–143–3p targeted S100A11 and was negatively regulated by circFOXP1. miR–143–3p competitively bound circFOXP1. circFOXP1 regulated HTR-8/SVneo cell functions through the miR–143–3p/S100A11 axis.
Wu H, et al. [73]2022Maternal bloodGDM vs. uncomplicatedCirc_102682 was decreased in GDM blood samples. circRNA_102682 was significantly correlated with triglycerides, APOA1, APOB, 1-h blood glucose in the serum of GDM patients.
She W, et al. [74]2021Maternal plasmaGDM vs. uncomplicatedCircVEGFC regulates glucose metabolism—higher incidence of GDM in patients with high circVEGFC levels. Elevated circVEGFC levels in GDM plasma. High circVEGFC level group showed higher incidence rates of fetal malformation and hypertension.Correlation established but further mechanistic studies required to establish whether this is causative.
Zou H, et al. [75]2022Human placentaPE vs. uncomplicatedCirc_0037078 is upregulated in PE placentae. Knockdown of circ_0037078 increases trophoblast cell proliferation, migration, invasion and angiogenesis. Circ_0037078 also sponges miR-576-5p and increases IL1RAP expression.
Mao Q, et al. [76]2021Human placentaPE vs. uncomplicatedCirc_0032962 and PBX3 levels were decreased in PE placentae and miR-326 was elevated. Circ_0032962 knockdown suppressed cell proliferation ability, migration, invasion, and EMT in HTR-8/SVneo cells.
Wang L, et al. [77]2021Maternal bloodPE vs. uncomplicatedCirc_0051326 and HLA-G protein and mRNA were decreased in PE samples. There was a positive correlation between the expression of serum circ_0051326 with HLA-G mRNA.
Shu C, et al. [78]2021Human placentaPE vs. uncomplicatedSilencing circ_0008726 promoted cell migration and
EMT, while circ_0008726 overexpression suppressed these processes. Circ_0008726 sponged miR-345-3p to regulate RYBP expression. Circ_0008726 was negatively correlated with miR-345-3p and positively correlated with RYBP expression levels in PE placentae. Transfection of miR-345-3p mimic or RYBP knockdown counteracted the effects of circ_0008726 overexpression on cell migration and EMT.
Zhang Y, et al. [79]2021Human placentaPE vs. uncomplicatedIncreases in m6A-modified circRNAs are prevalent in PE placentae, with the main methylation changes occurring in the 3′ UTR and near the start codon. In PE, circPAPPA2 levels are decreased while m6A modification is increased. METTL14 increases circPAPPA2 m6A methylation and IGF2BP3 maintains circPAPPA2 stability.
Wang W, et al. [80]2021Human placentaPE vs. uncomplicatedIn PE placentae, circHIPK3 and KCMF1 were downregulated and miR-346 was upregulated. CircHIPK3 overexpression promotes trophoblast cell proliferation, migration and invasion, as well as decreasing cell cycle arrest and apoptosis. CircHIPK3 also targets miR-346 and regulates KCMF1 expression.
Li J, et al. [81]2022Human placentaPE vs. uncomplicatedCircPAPPA positively regulates trophoblast cell proliferation, migration and invasion, and causes apoptosis and cell cycle arrest, through the miR-3127-5p/HOXA7 axis.
Wang W, et al. [82]2022Human placentaPE vs. uncomplicatedCirc_0017068 was downregulated in PE placental samples.
Circ_0017068 overexpression promoted HTR-8/SVneo cell proliferation, cycle progression, and suppressed apoptosis while silencing of circ_0017068 exhibited opposite effects. Circ_0017068 targeted miR-330-5p to regulate XIAP expression, and through this regulated proliferation, cycle progression, and apoptosis.
Zhang Y, et al. [83]2021Human placenta and maternal plasmaPE vs. uncomplicatedPE predictive power was greatest when plasma sFLT1 and circBRAP levels were combined with uterine pulsatility index. CircBRAP was increased in PE placentae and may regulate miR-106b to decrease TEV-1 cell proliferation, invasion and apoptosis.
Yuan Y, et al. [84]2022Human placentaPE vs. uncomplicated2432 circRNAs were differentially expressed between PE and control tissues. hsa_circRNA_0001687/hsa-miR-532-3p/MMP14/AXL, hsa_circ_0001513/hsa-miR-188-5p/HMGCS1 and hsa_circ_0001513/hsa_circ_0001329/hsa-miR-760/MAP1LC3B axes may contribute to PE pathogenesis.The paper uses publicly available datasets instead of completing their own sequencing. As such, data quality cannot be ascertained. No luciferase assays to validate the potential axes listed.
Wang H, et al. [85]2022Pig ovariesAtretic vs. healthy folliclesCircSCL41A1 was elevated in healthy follicles compared with atretic follicles. miR-9820-5p competitively binds circSLC41A1 to regulate SRSF1. A circSLC41A1-miR-9820-5p-SRSF1 axis regulates follicular granulosa cell apoptosis.
Table 2. Key signalling pathways common to placental development and cancer, with references.
Table 2. Key signalling pathways common to placental development and cancer, with references.
Signalling PathwayPlacentaCancer
EGFR[95][96]
PI3K/Akt[97][98]
P53[99][100]
IGF[101,102][103,104]
TGF-β[105][106]
VEGF[107][108]
mTOR[109,110][111]
MAPK[112][113]
Jak/STAT[114][115]
Wnt/β-catenin[116][117]
NF-κB[118][119]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Arthurs, A.L.; Jankovic-Karasoulos, T.; Smith, M.D.; Roberts, C.T. Circular RNAs in Pregnancy and the Placenta. Int. J. Mol. Sci. 2022, 23, 4551. https://doi.org/10.3390/ijms23094551

AMA Style

Arthurs AL, Jankovic-Karasoulos T, Smith MD, Roberts CT. Circular RNAs in Pregnancy and the Placenta. International Journal of Molecular Sciences. 2022; 23(9):4551. https://doi.org/10.3390/ijms23094551

Chicago/Turabian Style

Arthurs, Anya L., Tanja Jankovic-Karasoulos, Melanie D. Smith, and Claire T. Roberts. 2022. "Circular RNAs in Pregnancy and the Placenta" International Journal of Molecular Sciences 23, no. 9: 4551. https://doi.org/10.3390/ijms23094551

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