Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

MicroRNA Profiles in Spontaneous Decidualized Menstrual Endometrium and Early Pregnancy Decidua with Successfully Implanted Embryos

  • Yu Wang ,

    Contributed equally to this work with: Yu Wang, Yang Lv

    Affiliations The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, China, Department of Gynecology and Obstetrics, Inner Mongolia Medical University affiliated hospital, Hohhot, China

  • Yang Lv ,

    Contributed equally to this work with: Yu Wang, Yang Lv

    Affiliation The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, China

  • Shujun Gao,

    Affiliation College of Life Sciences, Inner Mongolia University, Hohhot, China

  • Yuanyuan Zhang,

    Affiliation College of Life Sciences, Inner Mongolia University, Hohhot, China

  • Jiajia Sun,

    Affiliation The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, China

  • Chunling Gong,

    Affiliation The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, China

  • Xiujuan Chen ,

    chenxiujuan616@163.com (XJC); gpengli@imu.edu.cn (GPL)

    Affiliation Department of Gynecology and Obstetrics, Inner Mongolia Medical University affiliated hospital, Hohhot, China

  • Guangpeng Li

    chenxiujuan616@163.com (XJC); gpengli@imu.edu.cn (GPL)

    Affiliation The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, China

Abstract

To comparatively analyze the human microRNA (miRNA) profiles between spontaneous decidualized menstrual endometrium and early pregnancy decidua by an in-depth sequencing of miRNAs. The specific miRNAs expressed at conception might be involved in pregnancy establishment and expression of let-7f-5p and let-7g-5p was experimentally up-regulated or inhibited to assess the effect on the expression of IGF2BP-1 and IGF2R in vitro, respectively. Samples of endometria and deciduas were obtained from 25 women who suffered from tubal or male factor subfertility and from 35 early pregnant women who underwent pregnancy termination at 6–8 weeks gestation were irrespectively collected and comparatively analyzed by miRNA sequencing and differential expression of known and novel miRNAs was analyzed using bioinformatics. The 2042 miRNA expression was analyzed in the study and the differential expression of six miRNAs was validated by qRT-PCR. The expression of four miRNAs in decidua samples was down-regulated (miR-34c, miR-92a, miR-181a-5p, and miR-191), whereas the expression of miR-10a-5p and let-7f-5p was significantly up-regulated. The expression of IGF2BP-1 and IGF2R declined and increased with overexpression and inhibition of let-7f-5p and let-7g-5p, respectively. Changes in the expression of particular miRNAs might play a role in the physiology of decidualization following successful embryo implantation, ultimately resulting in continuous decidualization.

Introduction

MicroRNAs (miRNAs) are a class of small noncoding RNAs that regulate gene expression and play fundamental roles in many biological processes, including cell differentiation and proliferation. In general, miRNAs inhibit translation or induce mRNA degradation by binding to the 3’ untranslated region of the target mRNA. To date, over 2,000 human miRNAs have been registered in miRBase v19.0 [1, 2].

Abnormal miRNA expression is associated with a number of benign gynecologic conditions, malignancies, and fertility disorders [3]. The abundance of specific miRNAs as well as relative differences in expression level can be assessed through miRNA sequencing. Several independent studies have profiled miRNA expression in secretory and proliferative endometria [46]. Other studies have shown that differences in miRNA expression occur during the implantation window in the secretory endometrium of patients either with or without endometriosis [7, 8]. However, to date, no reports have described the miRNA profiles of the spontaneous decidualized menstrual endometrium and early pregnancy decidua.

Menstruation is triggered by a decline in the level of circulating progesterone and involves the periodic discharge of blood and mucosal tissue, which consists of the decidualizing superficial endometrium. In response to declining progesterone levels, spontaneous decidualization leads to menstrual shedding and subsequent regeneration of the endometrium. In contrast to the case with most mammals, embryo implantation is not required for decidualization of the human endometrium. Furthermore, menstruation and cyclic regeneration of the endometrium involve recruitment of stem cells, imparting an intrinsic capacity for adaptation of the decidual response to maximize reproductive success. Once initiated, decidualization proceeds through distinct phenotypic phases involving endometrial receptivity, embryo selection, and ultimately, resolution of pregnancy at pregnancy initiation [9, 10].

Based on our previous study [11], we hypothesized that various miRNAs might play a role in embryo implantation and decidualization. However, the profile of miRNA expression in the menstrual endometrium remains to be elucidated [12, 13]. Characterization of the miRNomes of the human early pregnancy decidua and menstrual endometrium via deep sequencing might thus identify those decidualization-associated miRNAs that are involved in embryo implantation [14, 15]. The aim of the present study, therefore, was to identify miRNAs that play an important role in embryo implantation by characterizing the expression of miRNAs in the human menstrual endometrium and early pregnancy decidua. The miRNA profile of the early pregnancy decidua was compared with that of the menstrual phase in order to obtain new insights into the roles of miRNAs at conception.

Materials and Methods

Tissues collection

Endometrial biopsies were performed on the second day of the menstrual cycle in 25 women who suffered from secondary infertility due to either tubal or male factors and who received treatment between October 2011 and April 2012 in the Reproductive Center of Inner Mongolia University Affiliated Hospital (Inner Mongolia, China). Participants ranged in age from 22 to 39 years (mean, 31 years). In addition, decidua tissue was collected from 35 women (mean age, 28.6 years; range, 19–39 years) with a normal pregnancy (based on ultrasound evidence of fetal heart activity) who underwent pregnancy termination at 6–8 weeks of gestation. Samples were collected for miRNA and associated target gene analysis. The decidua was separated from the trophoblast, and each tissue sample was immediately frozen in liquid nitrogen. Six normal-pregnancy deciduas and six menstrual endometria were collected for sequencing analysis from as previous mentioned. The women in each group from whom specimens were collected for sequencing did not differ significantly in age. The study was approved by the Institute Research Ethics Committee of the Inner Mongolia Medical University Affiliated Hospital, and written informed consent was obtained from all participants.

Small RNA library construction and sequencing

Total RNA (including miRNAs) was extracted using RNAiso plus (Takara, Shiga, Japan). All samples passed the RNA quality control criteria for Solexa sequencing (RNA 28S:18S ≥1.5, RNA integrity number ≥8). The sequencing procedure was as described previously [11]. Small RNAs perfectly matching the miRBase precursor sequence and those identified as known miRNA editing polymorphisms in the miRBase 19.0 database [16, 17] were annotated and calculated. Mireap software [18] was used to predict novel miRNAs, as described previously [11].

MiRNA target prediction and functional analysis

The function of an miRNA is ultimately determined by the genes that the miRNA targets and by the effect of the miRNA on the expression of those genes. The RNAhybrid software [19] was used to predict the target genes of the novel miRNAs identified in this study [20, 21].

In vitro overexpression and inhibition of let-7f-5p and let-7g-5p

Human embryonic kidney (HEK293T) cells and human endometrial stromal cells (hESCs) were obtained from the American Type Culture Collection and cultured in DMEM (Gibco, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (Gibco). Let-7f-5p and let-7g-5p mimics (dsRNA oligonucleotides) and inhibitors (single-stranded chemically modified oligonucleotides) (GenePharma, Shanghai, China) were used for the in vitro overexpression and inhibition of let-7f-5p and let-7g-5p activity, respectively in S1 Table. Cells were plated in 6-well plates and cultured to 50–70% confluence prior to transfection. Lipofectamine LTX (Invitrogen, Carlsbad, CA, USA) was used for transfection according to the manufacturer’s instructions. Cells were transfected with RNAs at a final concentration of 10 nM; negative controls for the miRNA mimics and inhibitors were transfected as matched controls.

Quantitative reverse transcription PCR

Total RNA (1 μg) from each sample was used for the reverse transcriptase (RT) reaction to generate cDNAs using a microRNA RT kit (Takara). Quantitative real-time PCR was performed (in triplicate) using a SYBR Green PCR kit (Takara) according to the manufacturer’s instructions with an ABI Prism 7300HT real-time PCR system (Thermo Fisher Scientific, MA, USA). The PCR primers used in the study are listed in S1 Table. The expression levels of miRNAs and the predicted target genes (IGF2BP-1 and IGF2R) were normalized to those of the U6 snRNA and ACTB, respectively. Relative expression was calculated using the 2-ΔΔCT method [22].

Statistical analysis

All data are presented as the mean ± SD. The statistical significance of differences between experimental groups was assessed using the Student’s t-test or nonparametric test, and a P-value <0.05 was considered indicative of statistical significance.

Results

Profiles of miRNA expression in human early pregnancy decidua and menstrual endometrium

A total of 2,042 known miRNAs were expressed in the human decidua and endometrium samples analyzed. In addition, 177 miRNAs were differentially expressed between groups, including 88 that were up-regulated and 89 that were down-regulated (S2 Table). Clean sequence reads for the abundantly expressed miRNAs (>10,000 transcripts per million [TPM]) were used for further analyses. The known miRNAs in both groups exhibited a similar distribution in each library, based on classification by TPM. In normal decidua, 96.82% of miRNAs were expressed at low levels (<10 TPM), 3.04% were expressed at moderate levels (10–10,000 TPM), and only 0.15% of miRNAs were abundantly expressed (>10,000 TPM). However, the abundantly expressed miRNAs accounted for 87.11% of all miRNA reads. In the terminated-pregnancy decidua, 97.01% of miRNAs were expressed at <10 TPM, 2.84% were expressed at 10–10,000 TPM, and only 0.15% of miRNAs were expressed at >10,000 TPM. The abundantly expressed miRNAs accounted for 87.6% of all miRNA reads.

The three most abundantly expressed miRNAs in both groups were hsa-let-7f-5p, hsa-miR-199a-3p, and hsa-miR-199b-3p, accounting for 73.28%, 6.91%, and 6.91% of the miRNome, respectively. The differential expression of six abundantly expressed miRNAs (miR-34c, miR-92a, miR-181a-5p, miR-191, miR-10a-5p, and let-7f-5p) was verified by qRT-PCR analysis of the 35 normal-pregnancy deciduas and 25 menstrual endometria (Fig 1). In addition, qPCR analysis confirmed the differential expression of four significantly down-regulated miRNAs and two significantly up-regulated miRNAs in the normal-pregnancy deciduas examined.

thumbnail
Fig 1. Validation of the differential expression of six known miRNAs by qPCR analysis.

The miRNA expression level was normalized to that of U6 snRNA. “*” represents P value <0.05, “**” represents P value <0.01, and “***” represents P value <0.001. All experiments were repeated with 3 replicates.

https://doi.org/10.1371/journal.pone.0143116.g001

Discovery of novel miRNAs

A total of 62 novel miRNAs were some identified in deciduas and some in the menstrual endometria (Table 1). The expression of four novel miRNAs (miR-1, miR-12, miR-18, and miR-110) was markedly altered in the decidua, but these miRNAs were expressed at low levels (data not shown).

thumbnail
Table 1. The 62 novel miRNAs expressed in the human decidua and menstrual endometrium.

Candidates with sequence overlap with known miRNAs (but having a distinct mature miRNA sequence: isomiRs) and sequence alignments of novel miRNA candidates with known miRNAs of other species are shown.

https://doi.org/10.1371/journal.pone.0143116.t001

Target gene prediction and pathway analysis

A total of 37,513 target genes were predicted using Mireap software. The majority of the target genes were found to be involved in cellular processes, regulation of biological processes, and metabolic processes. The pathways with the most genes targeted by the identified miRNAs as determined by KEGG analysis are shown in Fig 2.

thumbnail
Fig 2. Kyoto Encyclopedia of Genes and Genomes analysis indicating the number of miRNA-targeted genes and the various associated pathways.

Horizontal axis indicates the annotated signaling pathways identified, and the vertical axis indicates the number of genes in each pathway to which miRNAs were predicted to bind.

https://doi.org/10.1371/journal.pone.0143116.g002

Target gene binding-site prediction

A number of candidate miRNA-target genes involved in establishing and maintaining pregnancy were identified in previous studies [11, 23, 24]. These binding-site predictions from our analyses suggest that interactions between miRNAs and mRNAs are involved in the maintenance of early pregnancy (Fig 3).

thumbnail
Fig 3.

(a) Let-7f-5p was predicted to bind to a site on the IGF2BP-1 mRNA. (b) Let-7g-5p was predicted to bind to a site on the IGF-2R mRNA.

https://doi.org/10.1371/journal.pone.0143116.g003

Changes in IGF2BP-1 and IGF2R expression in vitro after overexpression or knockdown of let-7f-5p and let-7g-5p

To further investigate the effect of let-7 miRNA on decidualization, the expression of target genes was evaluated following overexpression or knockdown of the miRNAs mediated by corresponding miRNA mimics or inhibitors, respectively. Because miRNAs of the let-7 family were found to be abundantly expressed in decidua in a previous study [11], our in vitro experiments had focused solely on the effects of let-7f and let-7g-5p. In the present study, we found that the expression of IGF2BP-1 mRNA increased in HEK293T and hESCs cells transfected with let-7f-5p inhibitor and decreased in cells transfected with let-7f-5p mimic (Fig 4). Likewise, the expression of IGF2R mRNA increased in cells transfected with let-7g-5p inhibitor and decreased in cells transfected with let-7g-5p mimic (Fig 5).

thumbnail
Fig 4. IGF2BP-1 expression in vitro after overexpression or knockdown of let-7f-5p.

(a) Overexpression of let-7f-5p in HEK293T and hESCs cells. Cells were cultured and incubated with hsa-let-7f-5p mimic or negative control. Levels of let-7f-5p were determined using qPCR. (b) Knockdown of let-7f miRNA by transfection with hsa-let-7f-5p inhibitor. Cells were cultured and incubated with hsa-let-7f-5p inhibitor or negative control. Levels of let-7f-5p were determined using qPCR. (c-d) Expression of IGF2BP-1 mRNA, as determined using qPCR analysis. Experiments were replicated three times, and error bars represent standard error. **P<0.01, ***P<0.001.

https://doi.org/10.1371/journal.pone.0143116.g004

thumbnail
Fig 5. IGF2R expression in vitro after overexpression or knockdown of let-7g-5p.

(a) Overexpression of let-7g-5p in HEK293T and hESCs cells. Cells were cultured and incubated with let-7g-5p mimic or negative control. Levels of let-7g-5p were determined using qPCR. (b) Knockdown of let-7g-5p miRNA by transfection with hsa-let-7g-5p inhibitor. Cells were cultured and incubated with hsa-let-7g-5p inhibitor or negative control. Levels of let-7g-5p were determined using qPCR. (c-d) Expression of IGF2R mRNA, as determined using qPCR analysis. Experiments were replicated three times, and error bars represent standard error. *P<0.05, **P<0.01 ***P<0.001.

https://doi.org/10.1371/journal.pone.0143116.g005

Discussion

Decidualization involves the transformation of endometrial stromal fibroblasts into specialized secretory decidual cells that provide the nutritive and immuno-privileged matrix essential for embryo implantation and placental development. In contrast to the case with most mammals, embryo implantation is not required for decidualization of the human endometrium. A growing body of evidence suggests that specific miRNAs play roles in the development and progression of early pregnancy via the regulation of a wide variety of signaling pathways in the decidua, including those involved in mediating inflammation [25], local estrogen biosynthesis [26], progesterone resistance [8], endometrial stromal cell invasiveness [27], extracellular matrix remodeling [28], angiogenesis [29, 30], and epigenetic regulation [7].

In the present study, we investigated the miRNome of the normal-pregnancy decidua and menstrual endometrium (consisting of the decidualizing superficial endometrium, with decidualization triggered by a decline in the level of circulating progesterone). We identified 2,042 known miRNAs expressed in the samples of human decidua. A total of 177 miRNAs were differentially expressed between groups, 88 of which were up-regulated and 89 of which were down-regulated. In early pregnancy deciduas, 0.15% of the miRNAs were abundantly expressed (>10,000 TPM), and these miRNAs accounted for 87.11% of all miRNA reads. Similarly, in menstrual endometria, 0.15% of the miRNAs were abundantly expressed (>10,000 TPM), and these miRNAs accounted for 87.6% of all miRNA reads. The differential expression of six abundant miRNAs in deciduas from the terminated-pregnancy group was confirmed in further analyses.

In this study, we found that the expression of let-7f-5p was higher in the early pregnancy decidua than in the menstrual endometrium. However, little is currently known about the function and role of miRNAs of the let-7 family in the decidua, and no reports of the significance of let-7 involvement in embryo implantation have been published. Our previous study showed that let-7 family members were expressed abundantly in decidua and their down regulation in normal pregnancy might involve in decidualization comparing to the aborted decidua [11]. And some of let-7 family members also differentially expressed in decidua and menstrual endometrium. To investigate the function of let-7 family members, further study was performed. Hu et al [31] analyzed miRNA expression in mouse uterus using miRNA microarrays and showed that let-7a, let-7b, let-7c, let-7d, let-7e, let-7f-5p, let-7g-5p, and let-7i are highly expressed in IMs (inter-implantation sites). Lozoya [32] showed that let-7a miRNA expression was maximal in 6-week embryonic tissue, and abruptly dropped in the transition to week-7, to remain low in expression up to week-9.

These results suggest that let-7a may be involved in ectopic implantation. A study by Chan et al [33] showed that the expression of let-7b and let-7c is significantly lower in the choriodecidua compared with the placenta and amnion, whereas the expression of let-7d and let-7f is lower in the amnion relative to the choriodecidua and placenta. In addition, although miRNAs of the let-7 family are highly expressed in mature oocytes and zygotes, their expression declines in the two-cell stage [34]. Hence, the roles of let-7 miRNAs in decidualization at pregnancy initiation require further clarification.

Given their apparently crucial roles in establishing endometrium receptivity and in mediating events such as spontaneous miscarriage, we therefore sought to identify target genes by overexpression or knockdown of let-7 miRNA expression in vitro. We demonstrated for the first time that there is a significant inverse relationship between the expression of let-7 miRNAs and target genes. In mice, the Igf2r gene exhibits imprinted expression in post-implantation tissues coinciding with expression of Airn ncRNA[35], it has been demonstrated that bovine IGF2R also exhibits imprinted expression in post-implantation tissues[36]. The observed expression of bAIRN in only 11% of bovine conceptuses at Day 15 and 80% of conceptuses at Day 18 of gestation supports the suggestion that imprinted expression of IGF2R/AIRN is being established during the peri-implantation period, around the time of maternal recognition of pregnancy in cattle [37]. The expression of PDGFA, placenta growth factor, IGF2BP1 and IGF2BP3 were up-regulated in human trophectoderm cells in day 5 after human chorionic gonadotrophin administration, but their acceptors, such as PDGFRA and KDR were over-expressed in the receptive endometrium[38]. As IGF2BP1 and IGF2R [39] are known to be involved in pregnancy establishment and maintenance [40], we hypothesized that let-7 disrupts decidualization from a maternal aspect by down-regulating the expression of IGF2BP1 and IGF2R.

Conclusion

In summary, this study characterized the miRNomes of the human decidua and menstrual endometrium during pregnancy, providing new data that should prove useful in subsequent investigations of the roles played by miRNAs during decidualization from a maternal aspect. Our in vitro data reveal an inverse relationship between the expression of the abundant let-7 miRNAs and their target genes, suggesting that miRNAs are involved in mediating endometrial receptivity and decidualization.

Supporting Information

S1 Table. Description of miRNA primers used for real-time RT-PCR.

https://doi.org/10.1371/journal.pone.0143116.s001

(DOCX)

S2 Table. 177 Differentially expressed miRNAs in the human menstrual endometrium and early pregnancy decidua.

https://doi.org/10.1371/journal.pone.0143116.s002

(XLSX)

Acknowledgments

This work was supported by a Ministry of Science and Technology of China 973 Basic Science Project Grant (2012CB722306), the National Natural Science Foundation of China(81560260), the Inner Mongolia Natural Science Foundation of China (2014MS0346, 2015MS08125), the MERCK CREAT Project (2014), and the Open Research Fund of the State Key Laboratory of Inner Mongolia University (20130902).

Author Contributions

Conceived and designed the experiments: YW YL GPL. Performed the experiments: YW YL SJG YYZ CLG JJS. Analyzed the data: YW YL GPL. Contributed reagents/materials/analysis tools: XJC GPL. Wrote the paper: YW YL GPL.

References

  1. 1. Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. (2006) miRBase: microRNA sequences, targets and gene nomenclature. Nucleic acids research 34:D140–4. pmid:16381832.
  2. 2. Kozomara A, Griffiths-Jones S. (2011) miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic acids research 39:D152–7. pmid:21037258.
  3. 3. Siristatidis C, Vogiatzi P, Brachnis N, Liassidou A, Iliodromiti Z, Bettocchi S, et al. (2015) Review: MicroRNAs in assisted reproduction and their potential role in IVF failure. In Vivo 29:169–75. pmid:25792643.
  4. 4. Dior UP, Kogan L, Chill HH, Eizenberg N, Simon A, Revel A. (2014) Emerging roles of microRNA in the embryo-endometrium cross talk. Seminars in reproductive medicine 32:402–9. pmid:24959822.
  5. 5. Wessels JM, Edwards AK, Khalaj K, Kridli RT, Bidarimath M, Tayade C. (2013) The microRNAome of pregnancy: deciphering miRNA networks at the maternal-fetal interface. PloS one 8:e72264. pmid:24278102.
  6. 6. Kuokkanen S, Chen B, Ojalvo L, Benard L, Santoro N, Pollard JW. (2010) Genomic profiling of microRNAs and messenger RNAs reveals hormonal regulation in microRNA expression in human endometrium. Biology of reproduction 82:791–801. pmid:19864316.
  7. 7. Filigheddu N, Gregnanin I, Porporato PE, Surico D, Perego B, Galli L, et al. (2010) Differential expression of microRNAs between eutopic and ectopic endometrium in ovarian endometriosis. Journal of biomedicine & biotechnology 2010:369549. pmid:20300586.
  8. 8. Burney RO, Hamilton AE, Aghajanova L, Vo KC, Nezhat CN, Lessey BA, et al. (2009) MicroRNA expression profiling of eutopic secretory endometrium in women with versus without endometriosis. Molecular human reproduction 15:625–31. pmid:19692421.
  9. 9. Miravet-Valenciano JA, Rincon-Bertolin A, Vilella F, Simon C. (2015) Understanding and improving endometrial receptivity. Current opinion in obstetrics & gynecology 27:187–92. pmid:25827647.
  10. 10. Basile N, Caiazzo M, Meseguer M. (2015) What does morphokinetics add to embryo selection and in-vitro fertilization outcomes? Current opinion in obstetrics & gynecology 27:193–200. pmid:25699478.
  11. 11. Wang Y, Lv Y, Wang L, Gong C, Sun J, Chen X, et al. (2015) MicroRNAome in decidua: a new approach to assess the maintenance of pregnancy. Fertility and sterility 103:980–9 e6. pmid:25747134.
  12. 12. Carrington B, Sacks G, Regan L. (2005) Recurrent miscarriage: pathophysiology and outcome. Current opinion in obstetrics & gynecology 17:591–7. pmid:16258340.
  13. 13. Rai R, Regan L. (2006) Recurrent miscarriage. Lancet 368:601–11. pmid:16905025.
  14. 14. Altmae S, Martinez-Conejero JA, Esteban FJ, Ruiz-Alonso M, Stavreus-Evers A, Horcajadas JA, et al. (2013) MicroRNAs miR-30b, miR-30d, and miR-494 regulate human endometrial receptivity. Reprod Sci 20:308–17. pmid:22902743.
  15. 15. Gu Y, Sun J, Groome LJ, Wang Y. (2013) Differential miRNA expression profiles between the first and third trimester human placentas. American journal of physiology Endocrinology and metabolism 304:E836–43. pmid:23443922.
  16. 16. Ebhardt HA, Tsang HH, Dai DC, Liu Y, Bostan B, Fahlman RP. (2009) Meta-analysis of small RNA-sequencing errors reveals ubiquitous post-transcriptional RNA modifications. Nucleic acids research 37:2461–70. pmid:19255090.
  17. 17. Wu H, Ye C, Ramirez D, Manjunath N. (2009) Alternative processing of primary microRNA transcripts by Drosha generates 5' end variation of mature microRNA. PloS one 4:e7566. pmid:19859542.
  18. 18. Kwak PB, Wang QQ, Chen XS, Qiu CX, Yang ZM. (2009) Enrichment of a set of microRNAs during the cotton fiber development. BMC genomics 10:457. pmid:19788742.
  19. 19. Kruger J, Rehmsmeier M. (2006) RNAhybrid: microRNA target prediction easy, fast and flexible. Nucleic acids research 34:W451–4. pmid:16845047.
  20. 20. Allen E, Xie Z, Gustafson AM, Carrington JC. (2005) microRNA-directed phasing during trans-acting siRNA biogenesis in plants. Cell 121:207–21. pmid:15851028.
  21. 21. Schwab R, Palatnik JF, Riester M, Schommer C, Schmid M, Weigel D. (2005) Specific effects of microRNAs on the plant transcriptome. Developmental cell 8:517–27. pmid:15809034.
  22. 22. Livak KJ, Schmittgen TD. (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25:402–8. pmid:11846609.
  23. 23. Leach RE, Jessmon P, Coutifaris C, Kruger M, Myers ER, Ali-Fehmi R, et al. (2012) High throughput, cell type-specific analysis of key proteins in human endometrial biopsies of women from fertile and infertile couples. Hum Reprod 27:814–28. pmid:22215622.
  24. 24. Maslova MA, Smol'nikova VY, Savilova AM, Burmenskaya OV, Bystritskii AA, Tabolova VK, et al. (2015) Profiles of mRNA expression for genes involved in implantation, early and middle phases of secretion stage in human endometrium. Bulletin of experimental biology and medicine 158:781–4. pmid:25894777.
  25. 25. Lin SC, Wang CC, Wu MH, Yang SH, Li YH, Tsai SJ. (2012) Hypoxia-induced microRNA-20a expression increases ERK phosphorylation and angiogenic gene expression in endometriotic stromal cells. The Journal of clinical endocrinology and metabolism 97:E1515–23. pmid:22648654.
  26. 26. Toloubeydokhti T, Pan Q, Luo X, Bukulmez O, Chegini N. (2008) The expression and ovarian steroid regulation of endometrial micro-RNAs. Reprod Sci 15:993–1001. pmid:19088369.
  27. 27. Dai L, Gu L, Di W. (2012) MiR-199a attenuates endometrial stromal cell invasiveness through suppression of the IKKbeta/NF-kappaB pathway and reduced interleukin-8 expression. Molecular human reproduction 18:136–45. pmid:21989168.
  28. 28. Ohlsson Teague EM, Van der Hoek KH, Van der Hoek MB, Perry N, Wagaarachchi P, Robertson SA, et al. (2009) MicroRNA-regulated pathways associated with endometriosis. Mol Endocrinol 23:265–75. pmid:19074548.
  29. 29. Hawkins SM, Creighton CJ, Han DY, Zariff A, Anderson ML, Gunaratne PH, et al. (2011) Functional microRNA involved in endometriosis. Mol Endocrinol 25:821–32. pmid:21436257.
  30. 30. Ramon LA, Braza-Boils A, Gilabert-Estelles J, Gilabert J, Espana F, Chirivella M, et al. (2011) microRNAs expression in endometriosis and their relation to angiogenic factors. Hum Reprod 26:1082–90. pmid:21335415.
  31. 31. Hu SJ, Ren G, Liu JL, Zhao ZA, Yu YS, Su RW, et al. (2008) MicroRNA expression and regulation in mouse uterus during embryo implantation. The Journal of biological chemistry 283:23473–84. pmid:18556655.
  32. 32. Lozoya T, Dominguez F, Romero-Ruiz A, Steffani L, Martinez S, Monterde M, et al. (2014) The Lin28/Let-7 system in early human embryonic tissue and ectopic pregnancy. PloS one 9:e87698. pmid:24498170.
  33. 33. Chan HW, Lappas M, Yee SW, Vaswani K, Mitchell MD, Rice GE. (2013) The expression of the let-7 miRNAs and Lin28 signalling pathway in human term gestational tissues. Placenta 34:443–8. pmid:23545322.
  34. 34. Flemr M, Moravec M, Libova V, Sedlacek R, Svoboda P. (2014) Lin28a is dormant, functional, and dispensable during mouse oocyte-to-embryo transition. Biology of reproduction 90:131. pmid:24829024.
  35. 35. Sleutels F, Zwart R, Barlow DP. (2002) The non-coding Air RNA is required for silencing autosomal imprinted genes. Nature 415:810–3. pmid:11845212.
  36. 36. Long JE, Cai X. (2007) Igf-2r expression regulated by epigenetic modification and the locus of gene imprinting disrupted in cloned cattle. Gene 388:125–34. pmid:17150312.
  37. 37. Farmer WT, Farin PW, Piedrahita JA, Bischoff SR, Farin CE. (2013) Expression of antisense of insulin-like growth factor-2 receptor RNA non-coding (AIRN) during early gestation in cattle. Animal reproduction science 138:64–73. pmid:23473694.
  38. 38. Haouzi D, Dechaud H, Assou S, Monzo C, de Vos J, Hamamah S. (2011) Transcriptome analysis reveals dialogues between human trophectoderm and endometrial cells during the implantation period. Hum Reprod 26:1440–9. pmid:21427117.
  39. 39. Miese-Looy G, VDH MJ, Edwards AK, Lamarre J, Tayade C. (2012) Expression of insulin-like growth factor (IGF) family members in porcine pregnancy. The Journal of reproduction and development 58:51–60. pmid:21685713.
  40. 40. Fisher SJ, Giudice LC. (2011) SGK1: a fine balancing act for human pregnancy. Nature medicine 17:1348–9. pmid:22064410.