Skip to main content

The role of insulin-like growth factor 2 mRNA binding proteins in female reproductive pathophysiology

Abstract

Insulin-like growth factor 2 (IGF2) mRNA binding proteins (IMPs) family belongs to a highly conserved family of RNA-binding proteins (RBPs) and is responsible for regulating RNA processing including localization, translation and stability. Mammalian IMPs (IMP1-3) take part in development, metabolism and tumorigenesis, where they are believed to play a major role in cell growth, metabolism, migration and invasion. IMPs have been identified that are expressed in ovary, placenta and embryo. The up-to-date evidence suggest that IMPs are involved in folliculogenesis, oocyte maturation, embryogenesis, implantation, and placentation. The dysregulation of IMPs not only contributes to carcinogenesis but also disturbs the female reproduction, and may participate in the pathogenesis of reproductive diseases and obstetric syndromes, such as polycystic ovary syndrome (PCOS), pre-eclampsia (PE), gestational diabetes mellitus (GDM) and gynecological tumors. In this review, we summarize the role of IMPs in female reproductive pathophysiology, and hope to provide new insights into the identification of potential therapeutic targets.

Introduction

Insulin-like growth factor 2 (IGF2) mRNA binding proteins (IMPs or IGF2BPs) belong to a highly conserved family of RNA-binding proteins (RBPs), which function to regulate the localization, stability and translation of mRNA and fine-tune the physiological function of the proteins encoded [1]. Members of the IMP family include IMP1–3 in mammals; Vegetal-1 mRNA-binding protein (Vg1RBP/Vera) in Xenopus laevis; zipcode-binding protein 1 (ZBP1) in chickens; murine coding region determinant-binding protein (CRD-BP) in mice; and Drosophila IMP (dIMP) [2, 3]. IMP1 is an ortholog of CRD-BP and ZBP1, which are respectively involved in the prevention of c-MYC degradation and the regulation of β-actin mRNA localization [4, 5]. Vg1RBP/Vera, also known as K homology domain-containing protein overexpressed in cancer (KOC), and human IMP3 are also orthologs [6]. Homologs of human IMP2 have been identified as IGF2BP2a and IGF2BP2b in Danio rerio. Moreover, the p62 protein, a 62 kDa isoform generated by IMP2, has been identified as an autoantigen in human hepatocellular carcinoma [7]. Mammalian IMPs (IMP1–3) were originally discovered in studies of embryogenesis [8] and subsequently recognized as oncofetal proteins. IMPs play a critical role in many cancers [9,10,11] and metabolic diseases [12], due to their role in the regulation of cell processes including cell proliferation, metabolism, invasion and migration. Immunohistochemistry and western blot studies of adult female gonadal tissue report expression of IMP1–3 in the ovary and placenta [13]. Accumulating evidence has shown that IMPs are involved in many aspects of reproductive physiology such as oocyte development [14], ovulation [15], implantation [16] and placentation [17]. Recent studies have implicated the dysregulation of IMPs in polycystic ovary syndrome (PCOS) [18], pre-eclampsia (PE) [19,20,21] and gestational diabetes mellitus (GDM) [22], in addition to endometrial and ovarian cancers [23, 24]. There are few relevant reports systematically describing the function of IMPs in female reproductive physiology and pathology. Thus, we present the first comprehensive review of the roles of IMPs in female reproductive pathophysiology and provide insights into potential underlying etiological mechanisms of female reproductive disorders.

Overview of IMPs

The protein structures of the three identified human IMPs (IMP1-3) are highly similar in domain order, and share an overall amino acid sequence identity of 56% [25]. Their canonical structure contains two RNA recognition motifs (RRM) located in the N-terminal region and four nuclear ribonucleoprotein (hnRNP) K homology (KH) domains in the C-terminal region [8]. The RRMs may help to stabilize IMP-mRNA complexes (protein-RNA complexes), and the KH domains mainly contribute to the binding of RNA [26]. Structural analyses indicate that binding of the KH domains 3 and 4 to mRNA induces a conformational change in the transcript, and therefore helps RBPs to assemble higher-order complexes through sequence-specific interactions, providing evidence for their high binding affinity and specificity [8, 27]. The long half-life of IMP-mRNA complexes confers high stability [28]. Moreover, RNA-binding motifs permit IMPs to bind cooperatively with more than one RNA simultaneously, permitting dimerization and formation of stable complexes.

The function of IMPs in the regulation of RNA metabolism has been widely studied, including RNA localization, stability and translation. IMPs are found to be predominantly expressed in the cytoplasm where they form messenger ribonucleoprotein (mRNP) granules around the nucleus which function to transiently “lock” target mRNAs. IMP1 granules have been shown to be particularly prevalent in neurons and oocytes. Compositional analysis has identified the exon-junction components, CBP80 and poly(A)-binding protein in IMP1-containing mRNP granules; the presence of which represents pre-translational status. IMP1 has also been shown to mediate transport of β-actin mRNA to the leading edge of motile cells.

IMPs were originally discovered as mRNA-binding proteins capable of binding to the 5’ untranslated regions (5’UTR) of IGF2 [8]. The IGF2 gene generates numerous mRNAs with different 5’UTRs. The human IGF-2 gene is a complex transcription unit driven by four promoters. Each promoter directs transcription of a different RNA designated leader. Four designated leaders of IGF2 have different translational properties. IMP1-3 have been shown to regulate the translation of IGF2 through binding with the IGF2 mRNA designated leader 3 rather than IGF2 mRNA designated leader 4 in a rapamycin-sensitive manner. The co-translational phosphorylation of IMP1 is catalyzed by mTOR complex 2, which mediates initiation of IGF2-L3-luciferase mRNA translation by cap-independent internal ribosomal entry [29]. IMP2 may also be phosphorylated by the mTOR complex 1 to promote translation of IGF2 mRNA by internal ribosomal entry [30]. Knockout of IMP3 results in the inhibition of translation of IGF2 and reduction in the levels of both intracellular and secreted IGF2 [31]. The IMP-IGF2 pathway has been implicated in the pathogenesis of many diseases. For example in immune thrombocytopenia, inactivation of IMP1-IGF2 signaling, caused by overexpression of miR-98-5p, has been shown to repress the phosphatidylinositol 3-kinase (PI3K)/Akt pathway and play a role in the deficiency of mesenchymal stem cells [32]. In cases of hypoxic-ischemic brain injury, activation of IMP2-IGF2 signaling, mediated by RNA-binding motif protein 3, promotes neural stem cell and progenitor cell proliferation and differentiation in the sub-granular zone [33]. In chicken myoblasts, the inhibition of IMP3-IGF2 pathway elicited by let-7b repressed cell proliferation has been found to be involved in the pathogenesis of dwarfism. In the female reproductive system, IGF2 has been universally found in follicular fluid [34], granulosa cells (GC) [35] and theca cells [36], and plays a role in folliculogenesis [37] and embryogenesis [38]. IGF2 is mainly secreted from granulosa cells, and collaborates with IGF1 to stimulate steroid hormone synthesis and promote proliferation of GCs through acting on IGF1R [39]. IMPs may therefore contribute to the normal physiology and pathological conditions of the female reproductive system through the regulation of IGF2. Other mechanisms of action of IMPs in the female reproductive system are described below.

Distribution of IMPs in the ovary

Expression of all three IMPs have been identified in adult ovarian and placental tissue. The physiological expression of IMP1–3 exhibits a biphasic pattern during development. IMP1–3 first appears in the oocyte and persists from pre-fertilization to the stages of zygote and blastocyst during early embryogenesis, and is then found to decline until murine embryonic day 10.5–12.5, when expression increases again [13]. In the adult human ovary, immunohistochemical studies indicate that IMPs are expressed in the cytoplasm of resting and growing oocytes, as well as GCs of small and growing follicles, with strong immunostaining of IMP2 and weak immunostaining of IMP1and IMP3. In human fetal ovaries, strong immunostaining of IMP3 and weak immunostaining of IMP1 were restricted to developing follicles from 32 weeks gestation, while IMP2 immunostaining was ubiquitous. Evidence from immunohistochemical analysis suggests that IMP1–3 may therefore play several different roles in folliculogenesis.

IMPs in embryogenesis

After fertilization, the early embryo undergoes maternal-to-zygotic transition (MTZ), which is the stage during which regulation of development, previously controlled by maternal factors, changes to be under the control of new genetic products from the zygotic genome. The transition includes two distinct processes: first, the maternal information (mRNAs and proteins) is gradually cleared; second, zygotic transcription is activated [40]. Many maternal factors are present from oocyte to embryo and directly regulate early development in vertebrates, such as egg activation, embryonic cell division, and cytoskeleton assembly [41,42,43]. Maternally inherited mRNAs are stable during the first few hours of embryonic development, and then subsequently degrade [40]. During the MTZ, initiation of zygotic transcription, known as zygotic genome activation (ZGA), is mediated by maternally derived factors [44]. Although the exact timing of ZGA during MTZ remains uncertain, some genes involved in the process have been identified. IMP3 acts as an RNA stabilizer during early development, and depletion of maternal IMP3 destabilizes maternal mRNAs prior to MTZ and leads to severe developmental defects, including abnormal organization of the cytoskeleton and aberrant cell division [45]. Moreover, overexpression of IMP3 enhances the stability of its target maternal mRNAs, decelerates clearance of maternal mRNA, and inhibits MTZ, resulting in developmental delay [45, 46]. Thus, maintaining a balance of IMP3 is essential for normal early development. Deletion of IMP2 has been shown to cause early embryonic developmental arrest by targeting the 3’UTR of CCAR1 and RPS14 during ZGA [38]. IMP2 also enhances IGF2 mRNA stability and promotes IGF2 translation. In vitro experimentation suggests that supplementation of culture media of derived mouse embryos with IGF2 remarkably improves the proportion of embryos that successfully develop compared with a control group (IMP2♀+/♂+), but exerts no effect on IMP2−/−female-derived embryos (IMP2♀−/♂+). This effect of IGF2 has also been observed in human embryos [38]. The data suggest that IMP2 is involved in ZGA in part by regulating the stability and translation of IGF2.

Embryo implantation is a dynamic and complex reproductive process regulated by a series of molecular and cellular events with its ultimate success based on the established uterine receptivity [47, 48]. Both abnormal hereditary and epigenetic modifications can affect uterine receptivity and therefore disturb embryo implantation and result in spontaneous abortion [49, 50]. Whole-genome bisulphite sequencing and differentially methylated region (DMR) analysis showed that 21,391 DMRs were found to be hypomethylated in the receptive endometrium of goats on day 15 of gestation compared with the pre-receptive endometrium on day 5 of gestation. The methylation ratio of IMP2 has been found to be lower in the receptive endometrium of goats than in the pre-receptive endometrium, while methylation ratios of IMP3 and IGF1R genes are higher [33], which may be a compensatory response. Abnormal expression of IMPs in endometrium may affect embryo implantation by affecting uterine receptivity. Many members of the let-7 family have been found to inhibit IMP expression by binding to the 3’UTR of IMP mRNA in various cell types. IMP1 has been identified as a target of let-7a [51, 52], let-7b [53], let-7f [54] and let-7i [55]. IMP2 has been identified as a target of let-7a [56], let-7b [57, 58] and let-7i [55] and IMP3 has been identified as a target of let-7b [59] and let-7i [55]. Interestingly, the expression of members of the let-7 family has been found to be lower in endometrial samples with endometritis than in clinically healthy endometrium, in particular let-7e and let-7f that are repressed in both subclinical and clinical endometritis, which may disturb uterine homeostasis and affect uterine receptivity [58]. Therefore, IMPs may potentially play a role in establishing endometrial receptivity, mediated by members of the let-7 family.

Early placentation is critical to both perinatal fetal growth and postnatal fetal and maternal health. Transcriptome analysis performed during the implantation period revealed that there is overexpression of some placental growth factors, and IMP1 and IMP3 in human trophoblast ectoderm cells, as well as upregulation of corresponding receptors in the receptive endometrium [16], implying that these molecules play an important role in the early dialogue between the blastocyst and maternal endometrial cells. Cytotrophoblast cells are found to proliferate and differentiate into several different trophoblast lineages during early gestation [60] and dysregulated proliferation and differentiation of cytotrophoblast cells can result in severe developmental disorders including intrauterine growth retardation and perinatal death [61, 62]. In sheep, rapid proliferation of trophoblast cells occurs in the process of conceptus elongation, which is essential for the implantation, placentation, and successfully establishing pregnancy [63, 64]. Knockout of LIN28 (LIN28A and LIN28B) and the resultant increase in let-7 micro-RNAs (miRNAs) in day 16 trophectoderm cells have been shown to reduce the degree of elongation of the conceptus and down-regulate the expression of IMP1, IMP2, IMP3, HMGA1, ARID3B, and c-MYC. This results in impaired placentation, fetal growth restriction and reduced fertility in sheep [17]. Therefore it is clear that IMPs play a major role in many developmental processes including ZGA, early embryonic development, implantation and placentation (Fig. 1), but the specific regulatory mechanisms employed in humans require further exploration.

Fig. 1
figure 1

Abnormal expression of IMPs in the pathogenesis of PCOS

In mouse parthenogenetic embryos, the expression of IMP1 and IMP2 is perturbed compared to that in embryos which have been normally fertilized. As N6-methyladenosine (m6A) readers, IMPs recognize m6A modifications, which regulate splicing, translocation, stability, and translation. In mammals, the occurrence of abnormal m6A modifications during early development results in parthenogenetically activated oocytes that are not capable of developing to term [65]. Reduced expression of IMP1 elicited by miR-670 decreases the rates of cleavage and blastula formation in parthenogenetically activated embryos via the down-regulation of m6A expression [66]. This effect can be reversed by treatment with betaine or miR-670 inhibitor.

IMPs and female reproductive pathologies

IMPs and PCOS

Polycystic ovary syndrome (PCOS) is a common disorder of the ovary in women of reproductive age, and is characterized by hyperandrogenism, ovulatory dysfunction (anovulation or oligo-ovulation) and a polycystic ovarian morphology [67]. Most PCOS patients also suffer from metabolic abnormalities, such as insulin resistance and abdominal obesity which suggests potential adipocyte dysfunction, and these patients also have an increased lifetime risk of developing type 2 diabetes mellitus (T2DM) [68]. Multiple pathologic mechanisms have been found, however the exact etiology of PCOS is not fully understood.

In polycystic ovaries, early follicular growth is excessive, resulting in massive ovarian preantral follicles, which become arrested and do not proceed to develop a dominant follicle. PCOS is also usually accompanied by high rates of GC proliferation and low rates of GC apoptosis. GC is an important source of anti-Müllerian hormone (AMH), IGF-1 and IGF-2 in the ovary. Elevated AMH has an antagonistic effect against the action of follicle stimulating hormone (FSH) on GCs, and represses the conversion of androgens to estrogens, therefore aggravating the androgen excess found in PCOS. Large numbers of GCs associated with excessive amounts of small ovarian follicles, which have become arrested in part, account for the characteristic polycystic morphology of the ovaries in PCOS. The expression of IMPs in PCOS patients is controversial and IMP2 may play a more important regulatory role in ovaries than IMP1 and IMP3. A recent microarray analysis identified that IGF1R, IGF2R, IMP2, IGFBP2 and IGFBP7 were all found to be down-regulated in the cumulus cells from patients with PCOS compared with patients without PCOS [69]. In contrast, a recent study showed elevation of expression of IMP2 in GCs and ovarian tissue of PCOS patients compared with that of a control group [18]. This contradiction may be due to the small sample size of the earlier study. A genome-wide association study (GWAS) showed that the high mobility group AT hook 2 (HMGA2) gene may be a high risk gene for PCOS and the expression of HMGA2 in ovarian tissue and ovarian GCs of PCOS patients has been shown to be higher than that in a control group. IMP2 can be upregulated by overexpression of HMGA2 and can promote GC proliferation by targeting the 3’UTR of cyclin D2 (CCND2) and SERPINE1 mRNA binding protein 1 (SERBP1) mRNA [18]. In the ovary, IGF2, IGF1 and insulin are capable of synergistically stimulating androgen secretion from theca cells [70]. Abnormally elevated expression of IMP2 promotes the translation and stability of IGF2. Increased secretion of IGF2 stimulates the production of androgens from theca cells in ways that are synergistic with IGF1 and insulin, which helps to further explain the androgen excess found in PCOS patients.

Ovulatory dysfunction may be partially due to impaired oocyte maturation. In the zebrafish oocyte, IMP3 has been found to retard the progression of oocyte maturation by repressing translation of cyclin B1 mRNA [71]. Upregulation of IMP3 in bovine oocytes is related to a lower vitrification temperature, which may affect stress prevention and oocyte recovery [72], however temperature-dependent regulation of IMP3 in human oocytes remains unknown.

As mentioned earlier, many patients with PCOS have other concurrent metabolic disorders, such as insulin resistance, hyperinsulinemia, dyslipidemia and activation of proinflammatory factors, and these patients also have a long term risk of developing T2DM and obesity [68, 73, 74]. A case–control study suggested that variants of T2DM risk genes are significantly more frequently found in patients with PCOS than in healthy controls [75]. There is a transcription-regulatory mechanism involved in regulating insulin secretion and glucose metabolism. Conditional inactivation of IMP2 has been shown to result in impaired insulin secretion from the islet cells of mice [76]. Reduced expression of multiple miRNAs including let-7b-5p, miR-1-3p, miR-24-3p, miR-34a-5p, miR-98-5p, and miR-133a-3p, has been observed in the peripheral blood of T2DM patients compared with controls and expression levels appear to correlate with serum insulin levels, HbA1c levels and body mass index (BMI) [77]. These miRNAs target susceptibility genes for T2DM including CDKN2A, CDK5, IMP2, KCNQ1, and TSPAN8. These results suggest that the elevated expression of IMP2 observed in PCOS may also play a role in inducing insulin resistance in addition to the abnormal promotion of GC proliferation. The molecular mechanisms of IMP2 related to the regulation of metabolism in PCOS remain to be confirmed. IMP1 is a critical regulator of fat metabolism and adipogenesis, but whether IMP1 influences the metabolic abnormalities found in PCOS patients remains to be explored. Interestingly, a study reported that metformin was found to downregulate H19 via mediation of DNA methylation [78]. H19 is a long noncoding RNA, which harbors multiple let-7 binding sites and acts as a ‘sponge’ to prevent let-7 from inhibiting expression of target genes [79, 80]. The data suggest that metformin may repress IMPs by downregulating the H19/let-7 pathway of DNA methylation, which may be one of its therapeutic mechanisms. All in all, the antagonistic action of miRNAs and IMPs, as well as the regulation of IMPs on target genes are both essential for the physiological and pathological processes involved in PCOS. IMPs are likely to participate in the pathogenesis of PCOS in a variety of ways (Fig. 2).

Fig. 2
figure 2

The effect of IMPs on parthenogenetic activation, zygotic genomic activation, early embryonic development, implantation and placentation

IMPs and PE

Abnormal proliferation and differentiation of human placental trophoblasts result in abnormal trophoblast invasion and dysfunctional syncytialization, which are involved in the pathogenesis of PE [81]. As a serious pregnancy-related disease manifesting with hypertension and proteinuria, PE is one of the main conditions contributing to maternal mortality, perinatal death, preterm birth and intrauterine growth retardation [82]. The expression of miR-423-5p is increased in the placentas of pre-eclamptic patients and overexpression of miR-423-5p has been found to inhibit migration, invasion and proliferation as well as inducing apoptosis in HTR-8/SVneo cells via targeting IMP1 [21]. Elevated expression level of miR-181a-5p in both plasma and placenta of severe pre-eclamptic patients suppresses invasion and migration of HTR-8/SVneo cells by directly inhibiting IMP2 [20]. During normal pregnancy, IMP3 is highly expressed in human placental villi in the first trimester compared with the third trimester, particularly in cytotrophoblast cells and column trophoblast cells [19], therefore migration and invasion of trophoblast cells from the placenta in the first trimester are promoted compared with the third trimester [83]. Silencing of IMP3 notably represses the invasion and migration of first trimester human placental villi and decreases the mRNA level of IGF2 and CD44 in HTR8/SVneo cells [19]. IGF2 is reported to activate and stimulate the PI3K/AKT pathway and promote the migration of ovine trophoblast ectoderm cells [84, 85]. In addition, the phosphorylation of AKT promotes the metastasis of multiple tumor cells. Lower expression of IMP3 has been observed in the placentas of PE patients compared with placentas from healthy women [19]. Therefore, these studies suggest that down-regulation of IMP3 mediates abnormal invasion and migration of placental trophoblast cells, probably via IGF2-PI3K-AKT pathways, thus contributing to the development of PE.

IMPs and gestational diabetes mellitus (GDM)

Meta-analyses of candidate gene studies and GWAS have identified multiple genes which are reproducibly associated with GDM, including TCF7L2, GCK, KCNJ11, KCNQ1, CDKAL1, IMP2, MTNR1B, and IRS1. These genes are also associated with T2DM [86]. Among pregnant women, the presence of polymorphisms in IMP2 has been shown to be highly correlated with the occurrence of GDM [87]. Rs4402960 (IMP2) and rs1801278 (Gly972Arg, IRS1) have been shown to be significantly associated with a higher risk of GDM [88,89,90].

IMPs and gestational trophoblastic diseases

Gestational trophoblastic disease is caused by excess cellular proliferation of placental villous trophoblast cells, and includes a spectrum of cellular abnormalities including hydatidiform mole (HM), invasive mole, choriocarcinoma, and placental site trophoblast tumor. HMs can be divided into complete, partial and invasive HMs [91]. According to histopathological features, partial HMs appear to be similar to normal placenta, while complete and invasive HMs are more like choriocarcinoma with multiple abnormally expressed oncogenes [92]. Most previous studies have shown that high expression of IMP3 is positively correlated with the occurrence or progression of cancer. Conversely, IMP3 is strongly expressed in extravillous cytotrophoblasts in healthy placenta, with expression level gradually decreasing from partial HMs, to complete HMs to invasive HMs [93]. The data suggest that IMP3 expression is negatively correlated with the malignant potential of HMs, but the direct mechanisms require further exploration. In the study of choriocarcinoma, β-Catenin, as a critical mediator of the Wnt signaling pathway, promotes the proliferation of human choriocarcinoma cells via the LIN28B/Let-7a/IMP1 pathway which mediates inhibition of let-7a, promotes IMP1 expression and thereby promotes proliferation of JAR cells. High expression of LIN28B has been observed in choriocarcinoma tissue compared with normal placental villi. However, whether the expression of IMP1 in choriocarcinoma promotes cell proliferation needs to be further explored in clinical specimens [52].

IMPs and gynecological tumors

Overexpression of IMP3 is associated with an unfavorable overall survival of patients with epithelial ovarian carcinoma (EOC) [94], such as ovarian clear cell carcinoma (OCCC) [23, 95], ovarian serous carcinoma [96] and primary ovarian mucinous carcinoma [97]. Knockout of IMP3 inhibits cell proliferation, migration and invasion, and downregulates the translation of MMP-2 and MMP-9 in OCCC cells [98]. Elevated IMP1 levels have been observed in ovarian carcinoma compared with normal peritoneum [99]. IMP1 stabilizes c-MYC and β-TrCP1 MRNA transcripts and has been shown to promote cell proliferation in IGROV-1 ovarian carcinoma [100]. IMP1 enhances the phenotype of invasive tumor cells by reducing the down-regulation of its miRNA-regulated target MRNA [101]. The effect of IMP1 on cancer-derived cells is conserved, while the roles of IMP2 and IMP3 vary in a cell-dependent manner. Immunohistochemistry has revealed strong staining of IMP3 and IMP2 in both ovarian serous carcinoma and tubal cancers and therefore IMP3 and IMP2 may be useful as biomarkers of pelvic high-grade serous carcinoma [102, 103].

Uterine leiomyomas (ULM) is characterized by histological and molecular heterogeneity. The pathogenesis of ULM is partially a result of several driver gene mutations, such as MED12 mutation, HMGA2 overexpression, and biallelic FH inactivation [104, 105]. ULM with HMGA2 overexpression also has increased IMP2 expression, as well as higher levels of AKT signaling and mitogenic activity than other ULM types. HMGA2 has been shown to activate AKT signaling through upregulation of IMP2 in embryonic rhabdomyosarcoma [106]. Knockout of HMGA2 in ULM cells causes inactivation of AKT signaling and upregulation of p16 and p21, which eventually lead to cell arrest [107]. In uterine leiomyosarcoma, IMP3 is an independent marker of poor prognosis [108]. In addition, IMP3 was upregulated in HPV16-positive cervical cancer and precancerous tissues compared with normal tissues and facilitated aerobic glycolysis by stabilizing HK2 mRNA, consequently promoting the malignant phenotype in cervical cancer cells [109].

Endometrial cancer is a common gynecologic malignancy, which originates from the endometrium and is composed of heterogeneous populations of cells. Traditionally, it has been classified into two types, type I (estrogen-dependent) and type II (estrogen-independent) based on clinical and epidemiological features [110]. Pathologically, endometrioid carcinoma is the most common subtype of type I endometrial carcinoma, and type II generally includes serous carcinoma and clear cell carcinoma [111]. On the basis of immunohistochemistry, the key biomarkers that are useful in the differentiation of endometrioid and serous endometrial carcinomas are estrogen receptors, progesterone receptors, IMP3, p53, and p16 [52, 112].

Conclusions

In summary, IMPs are implicated in the physiology and pathology of the female reproductive system. As a biomarker, IMPs promise to be very useful in being able to monitor the healthy embryogenesis and oocyte maturation, and to predict the occurrence and progression of some female reproductive diseases (Table 1). The expression of IMP2 is increased in ovarian GCs of women with PCOS, while the expression of IMP1, IMP2 and IMP3 is decreased in the placental tissue of women with PE. There is an association between down-regulation of IMPs and some adverse pregnancy outcomes, however the exact mechanisms are not yet fully understood. Although the role of the IMP family in tumorigenesis has been extensively explored, the role in diseases of the female reproductive system is still under investigation, with many studies conducted in other species and few in human tissue. Study limitations including small sample sizes, inadequate measurement methods and lack of control for confounding factors preclude our ability to draw firm conclusions from the current evidence. More focused studies are needed to address these limitations in order to disentangle the functions of IMPs and to clarify the clinical value as a novel biomarker of female reproductive diseases.

Table 1 The role of IMPs in the embryogenesis, oocyte maturation, and female reproductive pathology

Availability of data and materials

Not applicable.

References

  1. Müller-McNicoll M, Neugebauer KM. How cells get the message: dynamic assembly and function of mRNA-protein complexes. Nat Rev Genet. 2013;14(4):275–87. https://doi.org/10.1038/nrg3434.

    Article  CAS  PubMed  Google Scholar 

  2. Nielsen FC, Nielsen J, Christiansen J. A family of IGF-II mRNA binding proteins (IMP) involved in RNA trafficking. Scandinavian journal of clinical and laboratory investigation Supplementum. 2001;234:93–9.

    Article  CAS  Google Scholar 

  3. Fic W, Faria C, Johnston D St. DrosophilaIMP regulates Kuzbanian to control the timing of Notch signalling in follicle cells. Development. 2019;146(2):dev168963. https://doi.org/10.1242/dev.168963.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Doyle GA, Betz NA, Leeds PF, Fleisig AJ, Prokipcak RD, Ross J. The c-myc coding region determinant-binding protein: a member of a family of KH domain RNA-binding proteins. Nucleic Acids Res. 1998;26(22):5036–44. https://doi.org/10.1093/nar/26.22.5036.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ross AF, Oleynikov Y, Kislauskis EH, Taneja KL, Singer RH. Characterization of a beta-actin mRNA zipcode-binding protein. Mol Cell Biol. 1997;17(4):2158–65. https://doi.org/10.1128/mcb.17.4.2158.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mueller-Pillasch F, Pohl B, Wilda M, et al. Expression of the highly conserved RNA binding protein KOC in embryogenesis. Mech Dev. 1999;88(1):95–9. https://doi.org/10.1016/s0925-4773(99)00160-4.

    Article  CAS  PubMed  Google Scholar 

  7. Zhang JY, Chan EK, Peng XX, Tan EM. A novel cytoplasmic protein with RNA-binding motifs is an autoantigen in human hepatocellular carcinoma. J Exp Med. 1999;189(7):1101–10. https://doi.org/10.1084/jem.189.7.1101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Nielsen J, Christiansen J, Lykke-Andersen J, Johnsen AH, Wewer UM, Nielsen FC. A family of insulin-like growth factor II mRNA-binding proteins represses translation in late development. Mol Cell Biol. 1999;19(2):1262–70. https://doi.org/10.1128/mcb.19.2.1262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hu X, Peng WX, Zhou H, et al. IGF2BP2 regulates DANCR by serving as an N6-methyladenosine reader. Cell Death Differ. 2020;27(6):1782–94. https://doi.org/10.1038/s41418-019-0461-z.

    Article  CAS  PubMed  Google Scholar 

  10. Hedegaard Jensen G, Mortensen MB, Klöppel G, Nielsen MFB, Nielsen O, Detlefsen S. Utility of pVHL, maspin, IMP3, S100P and Ki67 in the distinction of autoimmune pancreatitis from pancreatic ductal adenocarcinoma. Pathol Res Pract. 2020;216(5): 152925. https://doi.org/10.1016/j.prp.2020.152925.

    Article  CAS  PubMed  Google Scholar 

  11. Mancarella C, Scotlandi K. IGF2BP3 From Physiology to Cancer: Novel Discoveries, Unsolved Issues, and Future Perspectives. Front Cell Dev Biol. 2019;7:363. https://doi.org/10.3389/fcell.2019.00363.

    Article  PubMed  Google Scholar 

  12. Christiansen J, Kolte AM, Hansen TV, Nielsen FC. IGF2 mRNA-binding protein 2: biological function and putative role in type 2 diabetes. J Mol Endocrinol. 2009;43(5):187–95. https://doi.org/10.1677/jme-09-0016.

    Article  CAS  PubMed  Google Scholar 

  13. Hammer NA, Hansen TV, Byskov AG, et al. Expression of IGF-II mRNA-binding proteins (IMPs) in gonads and testicular cancer. Reproduction. 2005;130(2):203–12. https://doi.org/10.1530/rep.1.00664.

    Article  CAS  PubMed  Google Scholar 

  14. Raty M, Ketoja E, Pitkanen T, Ahola V, Kananen K, Peippo J. In vitro maturation supplements affect developmental competence of bovine cumulus-oocyte complexes and embryo quality after vitrification. Cryobiology. 2011;63(3):245–55. https://doi.org/10.1016/j.cryobiol.2011.09.134.

    Article  CAS  PubMed  Google Scholar 

  15. Drouilhet L, Mansanet C, Sarry J, et al. The highly prolific phenotype of Lacaune sheep is associated with an ectopic expression of the B4GALNT2 gene within the ovary. PLoS Genet. 2013;9(9): e1003809. https://doi.org/10.1371/journal.pgen.1003809.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Haouzi D, Dechaud H, Assou S, Monzo C, de Vos J, Hamamah S. Transcriptome analysis reveals dialogues between human trophectoderm and endometrial cells during the implantation period. Hum Reprod. 2011;26(6):1440–9. https://doi.org/10.1093/humrep/der075.

    Article  CAS  PubMed  Google Scholar 

  17. Azarova IE, Klyosova EY, Lazarenko VA, Konoplya AI, Polonikov AV. rs11927381 polymorphism and type 2 diabetes mellitus: contribution of smoking to the realization of susceptibility to the disease. Bull Exp Biol Med. 2020;168(3):313–6. https://doi.org/10.1007/s10517-020-04698-9.

    Article  CAS  PubMed  Google Scholar 

  18. Li M, Zhao H, Zhao SG, et al. The HMGA2-IMP2 pathway promotes granulosa cell proliferation in polycystic ovary syndrome. J Clin Endocrinol Metab. 2019;104(4):1049–59. https://doi.org/10.1210/jc.2018-00544.

    Article  PubMed  Google Scholar 

  19. Li W, Liu D, Chang W, et al. Role of IGF2BP3 in trophoblast cell invasion and migration. Cell Death Dis. 2014;5: e1025. https://doi.org/10.1038/cddis.2013.545.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wu L, Song WY, Xie Y, et al. miR-181a-5p suppresses invasion and migration of HTR-8/SVneo cells by directly targeting IGF2BP2. Cell Death Dis. 2018;9(2):16. https://doi.org/10.1038/s41419-017-0045-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Guo L, Liu Y, Guo Y, Yang Y, Chen B. MicroRNA-423-5p inhibits the progression of trophoblast cells via targeting IGF2BP1. Placenta. 2018;74:1–8. https://doi.org/10.1016/j.placenta.2018.12.003.

    Article  CAS  PubMed  Google Scholar 

  22. Wu L, Cui L, Tam WH, Ma RC, Wang CC. Genetic variants associated with gestational diabetes mellitus: a meta-analysis and subgroup analysis. Sci Rep. 2016;6:30539. https://doi.org/10.1038/srep30539.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Köbel M, Xu H, Bourne PA, et al. IGF2BP3 (IMP3) expression is a marker of unfavorable prognosis in ovarian carcinoma of clear cell subtype. Modern Pathol. 2009;22(3):469–75. https://doi.org/10.1038/modpathol.2008.206.

    Article  CAS  Google Scholar 

  24. Hiramatsu K, Yoshino K, Serada S, et al. Similar protein expression profiles of ovarian and endometrial high-grade serous carcinomas. Br J Cancer. 2016;114(5):554–61. https://doi.org/10.1038/bjc.2016.27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bell JL, Wächter K, Mühleck B, et al. Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs): post-transcriptional drivers of cancer progression? Cellular and molecular life sciences : CMLS. 2013;70(15):2657–75. https://doi.org/10.1007/s00018-012-1186-z.

  26. Wächter K, Köhn M, Stöhr N, Hüttelmaier S. Subcellular localization and RNP formation of IGF2BPs (IGF2 mRNA-binding proteins) is modulated by distinct RNA-binding domains. Biol Chem. 2013;394(8):1077–90. https://doi.org/10.1515/hsz-2013-0111.

    Article  CAS  PubMed  Google Scholar 

  27. Chao JA, Patskovsky Y, Patel V, Levy M, Almo SC, Singer RH. ZBP1 recognition of beta-actin zipcode induces RNA looping. Genes Dev. 2010;24(2):148–58. https://doi.org/10.1101/gad.1862910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nielsen J, Kristensen MA, Willemoës M, Nielsen FC, Christiansen J. Sequential dimerization of human zipcode-binding protein IMP1 on RNA: a cooperative mechanism providing RNP stability. Nucleic Acids Res. 2004;32(14):4368–76. https://doi.org/10.1093/nar/gkh754.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Dai N, Christiansen J, Nielsen FC, Avruch J. mTOR complex 2 phosphorylates IMP1 cotranslationally to promote IGF2 production and the proliferation of mouse embryonic fibroblasts. Genes Dev. 2013;27(3):301–12. https://doi.org/10.1101/gad.209130.112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Dai N, Rapley J, Angel M, Yanik MF, Blower MD, Avruch J. mTOR phosphorylates IMP2 to promote IGF2 mRNA translation by internal ribosomal entry. Genes Dev. 2011;25(11):1159–72. https://doi.org/10.1101/gad.2042311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Liao B, Hu Y, Herrick DJ, Brewer G. The RNA-binding protein IMP-3 is a translational activator of insulin-like growth factor II leader-3 mRNA during proliferation of human K562 leukemia cells. J Biol Chem. 2005;280(18):18517–24. https://doi.org/10.1074/jbc.M500270200.

    Article  CAS  PubMed  Google Scholar 

  32. Wang Y, Zhang J, Su Y, et al. miRNA-98-5p Targeting IGF2BP1 Induces Mesenchymal Stem Cell Apoptosis by Modulating PI3K/Akt and p53 in Immune Thrombocytopenia. Molecular therapy Nucleic acids. 2020;20:764–76. https://doi.org/10.1016/j.omtn.2020.04.013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhu X, Yan J, Bregere C, et al. RBM3 promotes neurogenesis in a niche-dependent manner via IMP2-IGF2 signaling pathway after hypoxic-ischemic brain injury. Nat Commun. 2019;10(1):3983. https://doi.org/10.1038/s41467-019-11870-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Monget P, Monniaux D, Pisselet C, Durand P. Changes in insulin-like growth factor-I (IGF-I), IGF-II, and their binding proteins during growth and atresia of ovine ovarian follicles. Endocrinology. 1993;132(4):1438–46. https://doi.org/10.1210/endo.132.4.7681760.

    Article  CAS  PubMed  Google Scholar 

  35. Zhou J, Adesanya OO, Vatzias G, Hammond JM, Bondy CA. Selective expression of insulin-like growth factor system components during porcine ovary follicular selection. Endocrinology. 1996;137(11):4893–901. https://doi.org/10.1210/endo.137.11.8895362.

    Article  CAS  PubMed  Google Scholar 

  36. Perks CM, Denning-Kendall PA, Gilmour RS, Wathes DC. Localization of messenger ribonucleic acids for insulin-like growth factor I (IGF-I), IGF-II, and the type 1 IGF receptor in the ovine ovary throughout the estrous cycle. Endocrinology. 1995;136(12):5266–73. https://doi.org/10.1210/endo.136.12.7588270.

    Article  CAS  PubMed  Google Scholar 

  37. Monget P, Fabre S, Mulsant P, et al. Regulation of ovarian folliculogenesis by IGF and BMP system in domestic animals. Domest Anim Endocrinol. 2002;23:139–54. https://doi.org/10.1016/s0739-7240(02)00152-2.

    Article  CAS  PubMed  Google Scholar 

  38. Liu HB, Muhammad T, Guo Y, et al. RNA-Binding Protein IGF2BP2/IMP2 is a Critical Maternal Activator in Early Zygotic Genome Activation. Adv Sci (Weigh). 2019;6(15):1900295. https://doi.org/10.1002/advs.201900295.

  39. Spicer LJ, Aad PY. Insulin-like growth factor (IGF) 2 stimulates steroidogenesis and mitosis of bovine granulosa cells through the IGF1 receptor: role of follicle-stimulating hormone and IGF2 receptor. Biol Reprod. 2007;77(1):18–27. https://doi.org/10.1095/biolreprod.106.058230.

    Article  CAS  PubMed  Google Scholar 

  40. Tadros W, Lipshitz HD. The maternal-to-zygotic transition: a play in two acts. Development (Cambridge, England). 2009;136(18):3033–42. https://doi.org/10.1242/dev.033183.

    Article  CAS  Google Scholar 

  41. Laver JD, Marsolais AJ, Smibert CA, Lipshitz HD. Regulation and Function of Maternal Gene Products During the Maternal-to-Zygotic Transition in Drosophila. Curr Top Dev Biol. 2015;113:43–84. https://doi.org/10.1016/bs.ctdb.2015.06.007.

    Article  CAS  PubMed  Google Scholar 

  42. Wagner DS, Dosch R, Mintzer KA, Wiemelt AP, Mullins MC. Maternal control of development at the midblastula transition and beyond: mutants from the zebrafish II. Dev Cell. 2004;6(6):781–90. https://doi.org/10.1016/j.devcel.2004.04.001.

    Article  CAS  PubMed  Google Scholar 

  43. Eno C, Solanki B, Pelegri F. aura (mid1ip1l) regulates the cytoskeleton at the zebrafish egg-to-embryo transition. Development (Cambridge, England). 2016;143(9):1585–99. https://doi.org/10.1242/dev.130591.

    Article  CAS  Google Scholar 

  44. Hamatani T, Ko MSh, Yamada M, et al. Global gene expression profiling of preimplantation embryos. Hum Cell. 2006;19(3):98–117. https://doi.org/10.1111/j.1749-0774.2006.00018.x.

    Article  PubMed  Google Scholar 

  45. Ren F, Lin Q, Gong G, et al. Igf2bp3 maintains maternal RNA stability and ensures early embryo development in zebrafish. Communications biology. 2020;3(1):94. https://doi.org/10.1038/s42003-020-0827-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhao BS, Wang X, Beadell AV, et al. mA-dependent maternal mRNA clearance facilitates zebrafish maternal-to-zygotic transition. Nature. 2017;542(7642):475–8. https://doi.org/10.1038/nature21355.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hirate Y, Suzuki H, Kawasumi M, et al. Mouse Sox17 haploinsufficiency leads to female subfertility due to impaired implantation. Sci Rep. 2016;6:24171. https://doi.org/10.1038/srep24171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yen CF, Liao SK, Huang SJ, et al. Decreased Endometrial Expression of Leukemia Inhibitory Factor Receptor Disrupts the STAT3 Signaling in Adenomyosis During the Implantation Window. Reproductive sciences (Thousand Oaks, Calif). 2017;24(8):1176–86. https://doi.org/10.1177/1933719116681515.

    Article  CAS  Google Scholar 

  49. Silveira CO, Rezende CP, Ferreira MC, Del Puerto HL, Reis FM. Implantation Failure Is Associated With Increased α-Inhibin and β-Glycan Gene Expression in Secretory Phase Endometrium: Nested Case-Control Study of Infertile Women Undergoing IVF/Fresh Embryo Transfer. Reproductive sciences (Thousand Oaks, Calif). 2017;24(5):720–5. https://doi.org/10.1177/1933719116667490.

    Article  CAS  Google Scholar 

  50. Salilew-Wondim D, Ibrahim S, Gebremedhn S, et al. Clinical and subclinical endometritis induced alterations in bovine endometrial transcriptome and miRNome profile. BMC Genomics. 2016;17:218. https://doi.org/10.1186/s12864-016-2513-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhou J, Bi C, Ching YQ, et al. Inhibition of LIN28B impairs leukemia cell growth and metabolism in acute myeloid leukemia. J Hematol Oncol. 2017;10(1):138. https://doi.org/10.1186/s13045-017-0507-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wu J, Feng X, Du Y, et al. β-catenin/LIN28B promotes the proliferation of human choriocarcinoma cells via Let-7a repression. Acta Biochim Biophys Sin. 2019;51(5):455–62. https://doi.org/10.1093/abbs/gmz027.

    Article  CAS  PubMed  Google Scholar 

  53. Cheng M, Si Y, Niu Y, et al. High-throughput profiling of alpha interferon- and interleukin-28B-regulated microRNAs and identification of let-7s with anti-hepatitis C virus activity by targeting IGF2BP1. J Virol. 2013;87(17):9707–18. https://doi.org/10.1128/jvi.00802-13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wang Y, Chen X, Zhang Y, Song J. Potential proteins targeted by let-7f-5p in HeLa cells. Biosci Trends. 2017;11(3):363–5. https://doi.org/10.5582/bst.2017.01037.

    Article  CAS  PubMed  Google Scholar 

  55. Fawzy IO, Hamza MT, Hosny KA, Esmat G, Abdelaziz AI. Abrogating the interplay between IGF2BP1, 2 and 3 and IGF1R by let-7i arrests hepatocellular carcinoma growth. Growth factors (Chur, Switzerland). 2016;34:42–50. https://doi.org/10.3109/08977194.2016.1169532.

    Article  CAS  Google Scholar 

  56. Liu Y, Dong N, Miao J, Li C, Wang X, Ruan J. Lin28 promotes dental pulp cell proliferation via upregulation of cyclin-dependent proteins and interaction with let-7a/IGF2BP2 pathways. Biomed Pharmacother. 2019;113:108742. https://doi.org/10.1016/j.biopha.2019.108742.

  57. Schaeffer V, Hansen KM, Morris DR, LeBoeuf RC, Abrass CK. RNA-binding protein IGF2BP2/IMP2 is required for laminin-β2 mRNA translation and is modulated by glucose concentration. Am J Physiol Renal Physiol. 2012;303(1):F75-82. https://doi.org/10.1152/ajprenal.00185.2012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wu Y, Zhong JL, Hou N, et al. MicroRNA Let-7b inhibits keratinocyte migration in cutaneous wound healing by targeting IGF2BP2. Exp Dermatol. 2017;26(2):116–23. https://doi.org/10.1111/exd.13164.

    Article  CAS  PubMed  Google Scholar 

  59. Lin S, Luo W, Ye Y, et al. IGF2BP3Let-7b Regulates Myoblast Proliferation by Inhibiting Expression in Dwarf and Normal Chicken. Front Physiol. 2017;8:477. https://doi.org/10.3389/fphys.2017.00477.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Aplin JD. The cell biological basis of human implantation. Bailliere’s best practice & research Clinical obstetrics & gynaecology. 2000;14(5):757–64. https://doi.org/10.1053/beog.2000.0116.

  61. Red-Horse K, Zhou Y, Genbacev O, et al. Trophoblast differentiation during embryo implantation and formation of the maternal-fetal interface. J Clin Investig. 2004;114(6):744–54. https://doi.org/10.1172/jci22991.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Brosens I, Pijnenborg R, Vercruysse L, Romero R. The, “Great Obstetrical Syndromes” are associated with disorders of deep placentation. Am J Obstet Gynecol. 2011;204(3):193–201. https://doi.org/10.1016/j.ajog.2010.08.009.

  63. Spencer TE, Hansen TR. Implantation and Establishment of Pregnancy in Ruminants. Adv Anat Embryol Cell Biol. 2015;216:105–35. https://doi.org/10.1007/978-3-319-15856-3_7.

    Article  PubMed  Google Scholar 

  64. Wang J, Guillomot M, Hue I. Cellular organization of the trophoblastic epithelium in elongating conceptuses of ruminants. CR Biol. 2009;332(11):986–97. https://doi.org/10.1016/j.crvi.2009.09.004.

    Article  CAS  Google Scholar 

  65. Hao J, Xianfeng Y, Gao W, et al. The perturbed expression of m6A in parthenogenetic mouse embryos. Genet Mol Biol. 2019;42(3):666–70. https://doi.org/10.1590/1678-4685-gmb-2018-0212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Hao J, Hu H, Jiang Z, et al. microRNA-670 modulates Igf2bp1 expression to regulate RNA methylation in parthenogenetic mouse embryonic development. Sci Rep. 2020;10(1):4782. https://doi.org/10.1038/s41598-020-61816-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Lizneva D, Suturina L, Walker W, Brakta S, Gavrilova-Jordan L, Azziz R. Criteria, prevalence, and phenotypes of polycystic ovary syndrome. Fertil Steril. 2016;106(1):6–15. https://doi.org/10.1016/j.fertnstert.2016.05.003.

    Article  PubMed  Google Scholar 

  68. Stepto NK, Cassar S, Joham AE, et al. Women with polycystic ovary syndrome have intrinsic insulin resistance on euglycaemic-hyperinsulaemic clamp. Human reproduction (Oxford, England). 2013;28(3):777–84. https://doi.org/10.1093/humrep/des463.

    Article  CAS  Google Scholar 

  69. Haouzi D, Assou S, Monzo C, Vincens C, Dechaud H, Hamamah S. Altered gene expression profile in cumulus cells of mature MII oocytes from patients with polycystic ovary syndrome. Human reproduction (Oxford, England). 2012;27(12):3523–30. https://doi.org/10.1093/humrep/des325.

    Article  CAS  Google Scholar 

  70. Rosenfield RL, Ehrmann DA. The Pathogenesis of Polycystic Ovary Syndrome (PCOS): The Hypothesis of PCOS as Functional Ovarian Hyperandrogenism Revisited. Endocr Rev. 2016;37(5):467–520. https://doi.org/10.1210/er.2015-1104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Takahashi K, Kotani T, Katsu Y, Yamashita M. Possible involvement of insulin-like growth factor 2 mRNA-binding protein 3 in zebrafish oocyte maturation as a novel cyclin B1 mRNA-binding protein that represses the translation in immature oocytes. Biochem Biophys Res Commun. 2014;448(1):22–7. https://doi.org/10.1016/j.bbrc.2014.04.020.

    Article  CAS  PubMed  Google Scholar 

  72. Zhang F, Zhang ZY, Cai MD, et al. Effect of vitrification temperature and cryoprotectant concentrations on the mRNA transcriptome of bovine mature oocytes after vitrifying at immature stage. Theriogenology. 2020;148:225–35. https://doi.org/10.1016/j.theriogenology.2019.11.006.

    Article  CAS  PubMed  Google Scholar 

  73. Spritzer PM. Polycystic ovary syndrome: reviewing diagnosis and management of metabolic disturbances. Arq Bras Endocrinol Metabol. 2014;58(2):182–7. https://doi.org/10.1590/0004-2730000003051.

    Article  PubMed  Google Scholar 

  74. Delitala AP, Capobianco G, Delitala G, Cherchi PL, Dessole S. Polycystic ovary syndrome, adipose tissue and metabolic syndrome. Arch Gynecol Obstet. 2017;296(3):405–19. https://doi.org/10.1007/s00404-017-4429-2.

    Article  CAS  PubMed  Google Scholar 

  75. Ezzidi I, Mtiraoui N, Mohmmed Ali ME, Masoudi AA, Abu DF. Impact of variants on type-2 diabetes risk genes identified through genomewide association studies in polycystic ovary syndrome: a case-control study. J Genet. 2018;97(5):1213–23.

    Article  CAS  Google Scholar 

  76. Greenwald WW, Chiou J, Yan J, et al. Pancreatic islet chromatin accessibility and conformation reveals distal enhancer networks of type 2 diabetes risk. Nat Commun. 2019;10(1):2078. https://doi.org/10.1038/s41467-019-09975-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kokkinopoulou I, Maratou E, Mitrou P, et al. Decreased expression of microRNAs targeting type-2 diabetes susceptibility genes in peripheral blood of patients and predisposed individuals. Endocrine. 2019;66(2):226–39. https://doi.org/10.1007/s12020-019-02062-0.

    Article  CAS  PubMed  Google Scholar 

  78. Yan L, Zhou J, Gao Y, et al. Regulation of tumor cell migration and invasion by the H19/let-7 axis is antagonized by metformin-induced DNA methylation. Oncogene. 2015;34(23):3076–84. https://doi.org/10.1038/onc.2014.236.

    Article  CAS  PubMed  Google Scholar 

  79. Kallen AN, Zhou XB, Xu J, et al. The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol Cell. 2013;52(1):101–12. https://doi.org/10.1016/j.molcel.2013.08.027.

    Article  CAS  PubMed  Google Scholar 

  80. Fabian MR, Sonenberg N. The mechanics of miRNA-mediated gene silencing: a look under the hood of miRISC. Nat Struct Mol Biol. 2012;19(6):586–93. https://doi.org/10.1038/nsmb.2296.

    Article  CAS  PubMed  Google Scholar 

  81. Ji L, Brkić J, Liu M, Fu G, Peng C, Wang YL. Placental trophoblast cell differentiation: physiological regulation and pathological relevance to preeclampsia. Mol Aspects Med. 2013;34(5):981–1023. https://doi.org/10.1016/j.mam.2012.12.008.

    Article  CAS  PubMed  Google Scholar 

  82. Huppertz B. Placental origins of preeclampsia: challenging the current hypothesis. Hypertension. 2008;51(4):970–5. https://doi.org/10.1161/hypertensionaha.107.107607

  83. Hohn HP, Denker HW. Experimental modulation of cell-cell adhesion, invasiveness and differentiation in trophoblast cells. Cells Tissues Organs. 2002;172(3):218–36. https://doi.org/10.1159/000066965.

    Article  CAS  PubMed  Google Scholar 

  84. Kim J, Song G, Gao H, et al. Insulin-like growth factor II activates phosphatidylinositol 3-kinase-protooncogenic protein kinase 1 and mitogen-activated protein kinase cell Signaling pathways, and stimulates migration of ovine trophectoderm cells. Endocrinology. 2008;149(6):3085–94. https://doi.org/10.1210/en.2007-1367.

    Article  CAS  PubMed  Google Scholar 

  85. Ye Q, Cai W, Zheng Y, Evers BM, She QB. ERK and AKT signaling cooperate to translationally regulate survivin expression for metastatic progression of colorectal cancer. Oncogene. 2014;33(14):1828–39. https://doi.org/10.1038/onc.2013.122.

    Article  CAS  PubMed  Google Scholar 

  86. Lowe WL, Scholtens DM, Sandler V, Hayes MG. Genetics of Gestational Diabetes Mellitus and Maternal Metabolism. Curr DiabRep. 2016;16(2):15. https://doi.org/10.1007/s11892-015-0709-z.

    Article  CAS  Google Scholar 

  87. Chon SJ, Kim SY, Cho NR, Min DL, Hwang YJ, Mamura M. Association of variants in PPARγ2, IGF2BP2, and KCNQ1 with a susceptibility to gestational diabetes mellitus in a Korean population. Yonsei Med J. 2013;54(2):352–7. https://doi.org/10.3349/ymj.2013.54.2.352.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhang C, Bao W, Rong Y, et al. Genetic variants and the risk of gestational diabetes mellitus: a systematic review. Hum Reprod Update. 2013;19(4):376–90. https://doi.org/10.1093/humupd/dmt013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Cho YM, Kim TH, Lim S, et al. Type 2 diabetes-associated genetic variants discovered in the recent genome-wide association studies are related to gestational diabetes mellitus in the Korean population. Diabetologia. 2009;52(2):253–61. https://doi.org/10.1007/s00125-008-1196-4.

    Article  CAS  PubMed  Google Scholar 

  90. Wang Y, Nie M, Li W, et al. Association of six single nucleotide polymorphisms with gestational diabetes mellitus in a Chinese population. PLoS ONE. 2011;6(11): e26953. https://doi.org/10.1371/journal.pone.0026953.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Williams D, Hodgetts V, Gupta J. Recurrent hydatidiform moles. Eur J Obstet Gynecol Reprod Biol. 2010;150(1):3–7. https://doi.org/10.1016/j.ejogrb.2010.01.003.

    Article  PubMed  Google Scholar 

  92. Froeling FE, Seckl MJ. Gestational trophoblastic tumours: an update for 2014. Curr Oncol Rep. 2014;16(11):408. https://doi.org/10.1007/s11912-014-0408-y.

    Article  CAS  PubMed  Google Scholar 

  93. Wang J, Zhao M, Xiao J, Wu M, Song Y, Yin Y. E-Cadherin, CD44v6, and Insulin-Like Growth Factor-II mRNA-Binding Protein 3 Expressions in Different Stages of Hydatidiform Moles. J Biochem Mol Toxicol. 2016;30(9):455–61. https://doi.org/10.1002/jbt.21809.

    Article  CAS  PubMed  Google Scholar 

  94. Hsu KF, Shen MR, Huang YF, et al. Overexpression of the RNA-binding proteins Lin28B and IGF2BP3 (IMP3) is associated with chemoresistance and poor disease outcome in ovarian cancer. Br J Cancer. 2015;113(3):414–24. https://doi.org/10.1038/bjc.2015.254.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Bi R, Shen X, Zhang W, et al. Clear cell carcinomas of the ovary: a mono-institutional study of 73 cases in China with an analysis of the prognostic significance of clinicopathological parameters and IMP3 expression. Diagn Pathol. 2016;11:17. https://doi.org/10.1186/s13000-016-0467-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mohanty SK, Tiwari A, Singh C, et al. High-grade ovarian serous carcinomas: Significant correlation of histologic patterns with IMP3 and E-Cadherin predicting disease recurrence and survival. Ann Diagn Pathol. 2019;40:30–9. https://doi.org/10.1016/j.anndiagpath.2019.02.013.

    Article  PubMed  Google Scholar 

  97. Zhu Q, Qu Y, Zhang Q, et al. IMP3 is upregulated in primary ovarian mucinous carcinoma and promotes tumor progression. American journal of translational research. 2017;9(7):3387–98.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Liu H, Zeng Z, Afsharpad M, et al. Overexpression of IGF2BP3 as a Potential Oncogene in Ovarian Clear Cell Carcinoma. Front Oncol. 2019;9:1570. https://doi.org/10.3389/fonc.2019.01570.

    Article  PubMed  Google Scholar 

  99. Köbel M, Weidensdorfer D, Reinke C, et al. Expression of the RNA-binding protein IMP1 correlates with poor prognosis in ovarian carcinoma. Oncogene. 2007;26(54):7584–9. https://doi.org/10.1038/sj.onc.1210563.

    Article  CAS  PubMed  Google Scholar 

  100. Mahapatra L, Andruska N, Mao C, Le J, Shapiro DJ. A Novel IMP1 Inhibitor, BTYNB, Targets c-Myc and Inhibits Melanoma and Ovarian Cancer Cell Proliferation. Translational oncology. 2017;10(5):818–27. https://doi.org/10.1016/j.tranon.2017.07.008.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Müller S, Bley N, Glaß M, et al. IGF2BP1 enhances an aggressive tumor cell phenotype by impairing miRNA-directed downregulation of oncogenic factors. Nucleic Acids Res. 2018;46(12):6285–303. https://doi.org/10.1093/nar/gky229.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Hiramatsu K, Yoshino K, Serada S, et al. Similar protein expression profiles of ovarian and endometrial high-grade serous carcinomas. Br J Cancer. 2016;114(5):554–61. https://doi.org/10.1038/bjc.2016.27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Wang Y, Li L, Wang Y, et al. IMP3 as a cytoplasmic biomarker for early serous tubal carcinogenesis. Journal of experimental & clinical cancer research : CR. 2014;33:60. https://doi.org/10.1186/s13046-014-0060-2.

  104. Mäkinen N, Kämpjärvi K, Frizzell N, Bützow R, Vahteristo P. Characterization of MED12, HMGA2, and FH alterations reveals molecular variability in uterine smooth muscle tumors. Mol Cancer. 2017;16(1):101. https://doi.org/10.1186/s12943-017-0672-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Mehine M, Kaasinen E, Heinonen HR, et al. Integrated data analysis reveals uterine leiomyoma subtypes with distinct driver pathways and biomarkers. Proc Natl Acad Sci USA. 2016;113(5):1315–20. https://doi.org/10.1073/pnas.1518752113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Li Z, Zhang Y, Ramanujan K, Ma Y, Kirsch DG, Glass DJ. Oncogenic NRAS, required for pathogenesis of embryonic rhabdomyosarcoma, relies upon the HMGA2-IGF2BP2 pathway. Can Res. 2013;73(10):3041–50. https://doi.org/10.1158/0008-5472.can-12-3947.

    Article  CAS  Google Scholar 

  107. Xie J, Ubango J, Ban Y, Chakravarti D, Kim JJ, Wei JJ. Comparative analysis of AKT and the related biomarkers in uterine leiomyomas with MED12, HMGA2, and FH mutations. Genes Chromosom Cancer. 2018;57(10):485–94. https://doi.org/10.1002/gcc.22643.

    Article  CAS  PubMed  Google Scholar 

  108. Yasutake N, Ohishi Y, Taguchi K, et al. Insulin-like growth factor II messenger RNA-binding protein-3 is an independent prognostic factor in uterine leiomyosarcoma. Histopathology. 2018;72(5):739–48. https://doi.org/10.1111/his.13422.

    Article  PubMed  Google Scholar 

  109. Zhang Y, Zhao L, Yang S, et al. CircCDKN2B-AS1 interacts with IMP3 to stabilize hexokinase 2 mRNA and facilitate cervical squamous cell carcinoma aerobic glycolysis progression. Journal of experimental & clinical cancer research : CR. 2020;39(1):281. https://doi.org/10.1186/s13046-020-01793-7.

  110. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol. 1983;15(1):10–7. https://doi.org/10.1016/0090-8258(83)90111-7.

    Article  CAS  PubMed  Google Scholar 

  111. Böcker W. WHO classification of breast tumors and tumors of the female genital organs: pathology and genetics. Verh Dtsch Ges Pathol. 2002;86:116–9.

    PubMed  Google Scholar 

  112. Zaidi A, Gupta P, Gupta N, Rajwanshi A, Rai B, Gainder S. Role of Immunohistochemistry to Distinguish Grade 3 Endometrioid Carcinoma and Uterine Serous Carcinoma. Applied immunohistochemistry & molecular morphology : AIMM. 2020;28(1):42–8. https://doi.org/10.1097/pai.0000000000000720.

Download references

Acknowledgements

We would like to thank TopEdit (www.topeditsci.com) for English language editing of this manuscript.

Funding

Natural Science Foundation from Science and Technology Commission of Shanghai Municipality (grant No. 17ZR1403100 to Xue-Lian Li).

Author information

Authors and Affiliations

Authors

Contributions

X. X. performed the literature research, drafted the manuscript and created the figures. H. S. and X. L. helped to review the manuscript and discussed the completed manuscript. X. L. and J. Z. designed, supervised and reviewed the complete manuscript. The author(s) read and approved the final manuscript.

Corresponding authors

Correspondence to Jia-Rong Zhang or Xue-Lian Li.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that no competing interest exists.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, X., Shen, HR., Zhang, JR. et al. The role of insulin-like growth factor 2 mRNA binding proteins in female reproductive pathophysiology. Reprod Biol Endocrinol 20, 89 (2022). https://doi.org/10.1186/s12958-022-00960-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12958-022-00960-z

Keywords