Skip to main content
Log in

MiRNAs secreted by human blastocysts could be potential gene expression regulators during implantation

  • Original Article
  • Published:
Molecular Biology Reports Aims and scope Submit manuscript

Abstract

Background

Micro RNAs (miRNAs) are small non-coding RNAs known as essential regulators of cell–cell communication. Recent studies have revealed that miRNAs are secreted by a blastocyst in culture media. We hypothesized that endometrial epithelial cells take up embryo-derived miRNAs as well as other soluble factors and regulate their receptivity-related gene expression.

Methods and results

Blastocyst culture media (BCM) were collected from the individually cultured embryos, while human endometrial epithelial cells (HEECs) were collected from healthy fertile volunteers. To evaluate the effect of BCM on the endometrial receptivity gene expression, HEECs were co-cultured with implanted BCM, non-implanted BCM, and a control culture medium. After determining altered gene expression in the HEECs, the miRNAs-related genes through bioinformatics databases were identified and evaluated in the BCM. Co-culture of primary HEECs with BCM significantly stimulated the expression levels of VEGFA, HBEGF, HOXA10, and LIF in the implanted group compared with non-implanted and control groups. The fold changes of miR-195 significantly diminished in the implanted BCM group compared with the non-implanted BCM group. Reduced fold changes of miR‐29b, 145 and increased miR-223 were also observed in the implanted BCM group compared with the non-implanted ones.

Conclusion

miRNAs could function as potential gene expression regulators during implantation. These molecules are secreted by human blastocyst, taken up by endometrial epithelial cells, and cause a change in the endometrial function. We found that BCMs can be effective in implantation process by stimulating related receptivity gene expression.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in this published article.

References

  1. Tan Q et al (2020) MicroRNAs in small extracellular vesicles indicate successful embryo implantation during early pregnancy. Cells 9(3):645

    Article  CAS  Google Scholar 

  2. Quenby S, Brosens JJ (2013) Human implantation: a tale of mutual maternal and fetal attraction. Biol Reprod 88(3):1–2

    Article  Google Scholar 

  3. Luddi A et al (2021) Emerging role of embryo secretome in the paracrine communication at the implantation site: a proof of concept. Fertil Steril 115(4):1054–1062

    Article  CAS  Google Scholar 

  4. Haouzi D et al (2011) Transcriptome analysis reveals dialogues between human trophectoderm and endometrial cells during the implantation period. Hum Reprod 26(6):1440–1449

    Article  CAS  Google Scholar 

  5. Wang H et al (2018) Correlation of IL-1 and HB-EGF with endometrial receptivity. Exp Ther Med 16(6):5130–5136

    CAS  Google Scholar 

  6. Guo X et al (2021) Role of vascular endothelial growth factor (VEGF) in human embryo implantation: clinical implications. Biomolecules 11(2):253

    Article  CAS  Google Scholar 

  7. Bi Y et al (2022) HOXA10 improves endometrial receptivity by upregulating E-cadherin. Biol Reprod 106(5):992–999

    Article  Google Scholar 

  8. Whitby S, Zhou W, Dimitriadis E (2020) Alterations in epithelial cell polarity during endometrial receptivity: a systematic review. Front Endocrinol 11:596324

    Article  Google Scholar 

  9. Fukui Y et al (2022) Uterine receptivity is reflected by LIF expression in the Cervix. Reproductive Sci 29(5):1457–1462

    Article  CAS  Google Scholar 

  10. Edgell TA et al (2018) Assessment of potential biomarkers of pre-receptive and receptive endometrium in uterine fluid and a functional evaluation of the potential role of CSF3 in fertility. Cytokine 111:222–229

    Article  CAS  Google Scholar 

  11. Wu F et al (2018) Decreased MUC1 in endometrium is an independent receptivity marker in recurrent implantation failure during implantation window. Reprod Biol Endocrinol 16(1):1–7

    Article  Google Scholar 

  12. Rani V, Sengar RS (2022) Biogenesis and mechanisms of microRNA-mediated gene regulation. Biotechnol Bioeng 119(3):685–692

    Article  CAS  Google Scholar 

  13. Cuman C et al (2015) Human blastocyst secreted microRNA regulate endometrial epithelial cell adhesion. EBioMedicine 2(10):1528–1535

    Article  Google Scholar 

  14. Mouillet J-F et al (2015) MicroRNAs in placental health and disease. Am J Obstet Gynecol 213(4):S163–S172

    Article  CAS  Google Scholar 

  15. Valadi H et al (2007) Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 9(6):654–659

    Article  CAS  Google Scholar 

  16. Nahand JS et al (2020) Exosomal microRNAs: novel players in cervical cancer. Epigenomics 12(18):1651–1660

    Article  CAS  Google Scholar 

  17. Huang T et al (2019) The roles of extracellular vesicles in gastric cancer development, microenvironment, anti-cancer drug resistance, and therapy. Mol Cancer 18(1):1–11

    Article  CAS  Google Scholar 

  18. Berkhout RP et al (2020) High-quality human preimplantation embryos stimulate endometrial stromal cell migration via secretion of microRNA hsa-miR-320a. Hum Reprod 35(8):1797–1807

    Article  CAS  Google Scholar 

  19. Rosenbluth EM et al (2014) Human embryos secrete microRNAs into culture media—a potential biomarker for implantation. Fertil Steril 101(5):1493–1500

    Article  CAS  Google Scholar 

  20. Fang F et al (2021) MicroRNAs secreted by human embryos could be potential biomarkers for clinical outcomes of assisted reproductive technology. J Adv Res 31:25–34

    Article  CAS  Google Scholar 

  21. Capalbo A et al (2016) MicroRNAs in spent blastocyst culture medium are derived from trophectoderm cells and can be explored for human embryo reproductive competence assessment. Fertil Steril 105(1):225–235e3

    Article  CAS  Google Scholar 

  22. Marwood M et al (2009) Interleukin-11 and leukemia inhibitory factor regulate the adhesion of endometrial epithelial cells: implications in fertility regulation. Endocrinology 150(6):2915–2923

    Article  CAS  Google Scholar 

  23. Cuman C et al (2013) Preimplantation human blastocysts release factors that differentially alter human endometrial epithelial cell adhesion and gene expression relative to IVF success. Hum Reprod 28(5):1161–1171

    Article  CAS  Google Scholar 

  24. Sugino N et al (2002) Expression of vascular endothelial growth factor (VEGF) and its receptors in human endometrium throughout the menstrual cycle and in early pregnancy. Reprod Cambridge 123(3):379–387

    Article  CAS  Google Scholar 

  25. Torry DS et al (1996) Vascular endothelial growth factor expression in cycling human endometrium. Fertil Steril 66(1):72–80

    Article  CAS  Google Scholar 

  26. Hannan N et al (2011) Analysis of fertility-related soluble mediators in human uterine fluid identifies VEGF as a key regulator of embryo implantation. Endocrinology 152(12):4948–4956

    Article  CAS  Google Scholar 

  27. Sadekova ON et al (2015) Luteal phase defect is associated with impaired VEGF mRNA expression in the secretory phase endometrium. Reprod Biol 15(1):65–68

    Article  Google Scholar 

  28. Das SK et al (1994) Heparin-binding EGF-like growth factor gene is induced in the mouse uterus temporally by the blastocyst solely at the site of its apposition: a possible ligand for interaction with blastocyst EGF-receptor in implantation. Development 120(5):1071–1083

    Article  CAS  Google Scholar 

  29. Yoo HJ, Barlow DH, Mardon HJ (1997) Temporal and spatial regulation of expression of heparin-binding epidermal growth factor‐like growth factor in the human endometrium: a possible role in blastocyst implantation. Dev Genet 21(1):102–108

    Article  CAS  Google Scholar 

  30. Lessey BA et al (2002) Regulated expression of heparin-binding EGF‐like growth factor (HB‐EGF) in the human endometrium: a potential paracrine role during implantation. Mol Reprod Dev 62(4):446–455

    Article  CAS  Google Scholar 

  31. Satokata I, Benson G, Maas R (1995) Sexually dimorphic sterility phenotypes in Hoxa lO-deficient mice. Nature 374(6521):460–463

    Article  CAS  Google Scholar 

  32. Taylor HS et al (1998) HOXA10 is expressed in response to sex steroids at the time of implantation in the human endometrium. J Clin Investig 101(7):1379–1384

    Article  CAS  Google Scholar 

  33. Bagot CN, Kliman HJ, Taylor HS (2001) Maternal Hoxa10 is required for pinopod formation in the development of mouse uterine receptivity to embryo implantation. Dev Dyn 222(3):538–544

    Article  CAS  Google Scholar 

  34. Sakkas D et al (2003) A soluble molecule secreted by human blastocysts modulates regulation of HOXA10 expression in an epithelial endometrial cell line. Fertil Steril 80(5):1169–1174

    Article  Google Scholar 

  35. Shuya LL et al (2011) Leukemia inhibitory factor enhances endometrial stromal cell decidualization in humans and mice. PLoS ONE 6(9):e25288

    Article  CAS  Google Scholar 

  36. Song H et al (2000) Dysregulation of EGF family of growth factors and COX-2 in the uterus during the preattachment and attachment reactions of the blastocyst with the luminal epithelium correlates with implantation failure in LIF-deficient mice. Mol Endocrinol 14(8):1147–1161

    Article  CAS  Google Scholar 

  37. Dimitriadis E et al (2009) Local regulation of implantation at the human fetal-maternal interface. Int J Dev Biol 54(2–3):313–322

    Google Scholar 

  38. Aghajanova L (2004) Leukemia inhibitory factor and human embryo implantation. Ann N Y Acad Sci 1034(1):176–183

    Article  CAS  Google Scholar 

  39. Stewart CL et al (1992) Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature 359(6390):76–79

    Article  CAS  Google Scholar 

  40. Surveyor GA et al (1995) Expression and steroid hormonal control of Muc-1 in the mouse uterus. Endocrinology 136(8):3639–3647

    Article  CAS  Google Scholar 

  41. DeSouza MM et al (1998) Reduction of mucin-1 expression during the receptive phase in the rat uterus. Biol Reprod 58(6):1503–1507

    Article  CAS  Google Scholar 

  42. Bowen JA, Bazer FW, Burghardt RC (1996) Spatial and temporal analyses of integrin and Muc-1 expression in porcine uterine epithelium and trophectoderm in vivo. Biol Reprod 55(5):1098–1106

    Article  CAS  Google Scholar 

  43. Hild-Petito S et al (1996) Mucin (Muc-1) expression is differentially regulated in uterine luminal and glandular epithelia of the baboon (Papio anubis). Biol Reprod 54(5):939–947

    Article  CAS  Google Scholar 

  44. DeSouza MM et al (2000) MUC1/episialin: a critical barrier in the female reproductive tract. J Reprod Immunol 45(2):127–158

    Article  Google Scholar 

  45. Wang R et al (2013) MicroRNA-195 suppresses angiogenesis and metastasis of hepatocellular carcinoma by inhibiting the expression of VEGF, VAV2, and CDC42. Hepatology 58(2):642–653

    Article  CAS  Google Scholar 

  46. Caramuta S et al (2013) Clinical and functional impact of TARBP2 over-expression in adrenocortical carcinoma. Endocrine-related Cancer 20(4):551–564

    Article  CAS  Google Scholar 

  47. Zhu X-m et al (2009) Differential expression profile of microRNAs in human placentas from preeclamptic pregnancies vs normal pregnancies. Am J Obstet Gynecol 200(6):661. e1–661. e7

    Article  Google Scholar 

  48. Bai Y et al (2012) Downregulated miR-195 detected in preeclamptic placenta affects trophoblast cell invasion via modulating ActRIIA expression. PLoS ONE 7(6):e38875

    Article  CAS  Google Scholar 

  49. Bai X, Zheng C, Ren N (2019) Trophoblast proliferation, migration, and invasion is regulated by the miR-195/EGFR signaling pathway in recurrent spontaneous abortion patients. bioRxiv.  https://doi.org/10.1101/630590

    Article  Google Scholar 

  50. Choudhary A et al (2018) Identification of miR-29b targets using 3-cyanovinylcarbazole containing mimics. RNA 24(4):597–608

    Article  CAS  Google Scholar 

  51. Rarani FZ, Borhani F, Rashidi B (2018) Endometrial pinopode biomarkers: molecules and microRNAs. J Cell Physiol 233(12):9145–9158

    Article  CAS  Google Scholar 

  52. Wang X et al (2022) miRNA-29 aggravates myocardial infarction via inhibiting the PI3K/mTOR/HIF1α/VEGF pathway. Aging 14(7):3129

    Article  Google Scholar 

  53. Revel A et al (2011) MicroRNAs are associated with human embryo implantation defects. Hum Reprod 26(10):2830–2840

    Article  CAS  Google Scholar 

  54. Shekibi M et al (2022) Progesterone suppresses podocalyxin partly by up-regulating miR-145 and miR-199 in human endometrial epithelial cells to enhance receptivity in in-vitro models. Mol Hum Reprod. https://doi.org/10.1093/molehr/gaac034

    Article  Google Scholar 

  55. Dong X et al (2016) MicroRNA-223-3p suppresses leukemia inhibitory factor expression and pinopodes formation during embryo implantation in mice. Am J Transl Res 8(2):1155

    CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran for cooperating throughout the study. This study was financed by Iran University of Medical Sciences (Grant No. 97-01-208-33115).

Funding

This study was financed by Iran University of Medical Sciences (Grant No. 33115).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Fereshteh Mehraein or Fatemehsadat Amjadi.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (TIF 1168.1 kb)—Supplementary Fig. 1 Transferred blastocysts in 5th day.

11033_2022_8121_MOESM2_ESM.tif

Supplementary file2 (TIF 832.5 kb)—Supplementary Fig. 2 Primary endometrial glandular epithelium cell culture. Representative endometrial glandular epithelium at (A) ×100, (B) ×100, (C) ×200 and (D) ×200 magnification. Black arrows indicate glandular epithelium cells.

11033_2022_8121_MOESM3_ESM.tif

Supplementary file3 (TIF 171.6 kb)—Supplementary Fig. 3 Endometrial cell purity. Purified populations of endometrial epithelial cells were stained for the intracellular epithelial and mesenchymal markers cytokeratin and vimentin, respectively. Flow cytometry histograms present data in blue for cells stained with vimentin and cytokeratin, and in red for the cognate isotype control.

11033_2022_8121_MOESM4_ESM.tif

Supplementary file4 (TIF 53.2 kb)—Supplementary Fig. 4 Target genes and related microRNAs were selected according to the miRTargetLink 2.0.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Eivazi, S., Tanhaye Kalate Sabz, F., Amiri, S. et al. MiRNAs secreted by human blastocysts could be potential gene expression regulators during implantation. Mol Biol Rep 50, 1375–1383 (2023). https://doi.org/10.1007/s11033-022-08121-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11033-022-08121-6

Keywords

Navigation