Expression of exosomal microRNA in high-grade ovarian cancer and ovarian endometriotic cysts


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

Recently, exosomal microRNA has been considered a potential noninvasive biomarker of various proliferative processes, particularly affecting the female reproductive system. Objective: To investigate the potential of plasma exosomal microRNA expression in patients with serous ovarian cancer, extragenital endometriosis, and ovarian endometriotic cysts (OEC) as potential markers of these diseases. Materials and methods: The study included seven patients with histologically confirmed high-grade serous ovarian cancer, six women in a control group without this pathology ("Control-ovarian cancer"), six patients with OEC, and five women of reproductive age without this pathology ("Control-OEC"). The exosomes were isolated from the blood, and the composition of plasma-derived exosomal microRNAs was determined using next-generation sequencing. Results: Pairwise comparison of miRNA content in the groups "ovarian cancer" - "Control-ovarian cancer" and "OEC" - "Control-OEC" identified 22 and 13 differentially expressed miRNAs (DEM) with statistically significant 2-fold or more differences. Potential markers for ovarian cancer included miR-141-3p, -199a-5p, -200b-3p, -203a-3p, -224-5p, and -4488and for OEC, miR-92b-5p, -486-5p, -3184-3p, -4732-5p, and -4235p. Potential target genes were identified for DEM in each pair of groups. The intracellular signaling pathways most likely involved in the pathogenesis of ovarian cancer and OEC were searched based on their lists. Potential target genes of several signaling pathways regulated by one or more DEMs in ovarian cancer and OEC included AKT1, ATM, BARD1, BAX, BCL2, BRCA1, CASP3, CDK4, CHEK1, CHEK2, JAK1, MDM2, PLK1, PTEN, RB1, SMAD2, SMAD3, and TP53. Functional clustering of target genes showed their involvement in regulating signaling pathways of growth factors, cell cycle, apoptosis, and DNA repair. Conclusion: The detection of unique DEMs in plasma exosomes indicates the presence of specific changes in the microRNA profile characteristic of ovarian cancer and OEC. The listed exosomal microRNAs can be considered candidates for markers of the studied proliferative processes. Multiple interactions between the identified microRNA target genes indicate their significant contribution and joint involvement into proliferative processes in ovarian cancer and OEC.

Full Text

Restricted Access

About the authors

Mania V. Iurova

V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health of Russia; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)

Email: m_yurova@oparina4.ru
PhD. Student at the Chair of Obstetrics, Gynecology, Perinatology and Reproductology

Chupalav M. Eldarov

V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health of Russia

Email: ch_eldarov@oparina4.ru
PhD. (Bio), Senior Researcher at the Laboratory of Molecular Pathophysiolog

Mikhail Yu. Bobrov

V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health of Russia

Email: mbobr@mail.ru
PhD. (Chemistry), Head of the Laboratory of Molecular Pathophysiology

Grigory N. Khabas

V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health of Russia

Email: g_khabas@oparina4.ru
Ph.D., surgeon, oncologist, obstetrician-gynecologist, Head of the Department of Innovative Oncology and Gynecology

Stanislav V. Pavlovich

V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health of Russia; I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)

Email: s_pavbvich@oparina4.ru
PhD, Academic Secretary

References

  1. Siegel R.L., Miller K.D., Jemal A. Cancer statistics, 2017. CA Cancer J. Clin. 2017; 67(1):7-30. https://dx.doi.org/10.3322/caac.21387.2017; 67(1): 7-30.
  2. Serdar E. Bulun M.D. Endometriosis. N. Engl. J. Med. 2009; 360: 268-79. https://dx.doi.org/10.1056/NEJMra0804690.
  3. Павлович С.В., Юрова М.В., Мелкумян А.Г., Франкевич В.Е., Хабас Г.Н., Чаговец В.В. Биомаркеры при новообразованиях яичников: возможности, ограничения и перспективы применения у женщин репродуктивного возраста. Акушерство и гинекология. 2019; 11: 65-73. [Pavlovich S.V., Yurova M.V., Melkumyan A.G., Frankevich V.E., Chagovets V.V., Khabas G.N. Biomarkers in ovarian neoplasms: opportunities, limitations, and prospects for using in reproductive-aged women. Akusherstvo i Ginekologiya/Obstetrics and Gynecology. 2019; 11: 65-73. (in Russian)]. https://dx.doi.org/10.18565/aig.2019.11.65-73.
  4. Fassbender A., Burney R.O., O. D.F., D'Hooghe T., Giudice L. Update on biomarkers for the detection of endometriosis. Biomed. Res. Int. 2015; 2015: 130854. https://dx.doi.org/10.1155/2015/130854.
  5. Aznaurova Y.B., Zhumataev M.B., Roberts T.K., Aliper A.M., Zhavoronkov A.A. Molecular aspects of development and regulation of endometriosis. Reprod. Biol. Endocrinol. 2014 Jun 13; 12:50. https://dx.doi.org/10.1186/1477-7827-12-50.
  6. Cortez M.A., Bueso-Ramos C., Ferdin J., Lopez-Berestein G., Sood A.K., Calin G.A. MicroRNAs in body fluids-the mix of hormones and biomarkers. Nat. Rev. Clin. Oncol. 2011; 8(8): 467-77. https://dx.doi.org/10.1038/nrclinonc.2011.76.
  7. Mirzaei H., Gholamin S., Shahidsales S., Sahebkar A., Jaafari M.R., Mirzaei H.R. et al. MicroRNAs as potential diagnostic and prognostic biomarkers in melanoma. Eur. J. Cancer. 2016; 53: 25-32. https://dx.doi.org/10.1016/j.ejca.2015.10.009.
  8. Iorio M.V., Visone R., Di Leva G., Donati V., Petrocca F., Casalini P. et al. MicroRNA signatures in human ovarian cancer. Cancer Res. 2007; 67(18): 8699-707. https://dx.doi.org/10.1158/0008-5472.CAN-07-1936.
  9. Filigheddu N., Gregnanin I., Porporato P.E., Surico D., Perego B., Galli L. et al. Differential expression of micrornas between eutopic and ectopic endometrium in ovarian endometriosis. J. Biomed. Biotechnol. 2010; 2010: 369549. https://dx.doi.org/10.1155/2010/369549.
  10. Anfossi S., Babayan A., Pantel K., Calin G.A. Clinical utility of circulating non-coding RNAs - an update. Nat. Rev. Clin. Oncol. 2018; 15(9): 541-63. https://dx.doi.org/10.1038/s41571-018-0035-x.
  11. Farooqi A.A., Desai N.N., Qureshi M.Z., Librelotto D.R.N., Gasparri M.L., Bishayee A. et al. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol. Adv. 2018; 36(1): 328-34. https://dx.doi.org/10.1016/j.biotechadv.2017.12.010.
  12. Giannopoulou L., Zavridou M., Kasimir-Bauer S., Lianidou E.S. Liquid biopsy in ovarian cancer: the potential of circulating miRNAs and exosomes. Transl. Res. 2019; 205: 77-91. https://dx.doi.org/10.1016/j.trsl.2018.10.003.
  13. Aboutalebi H., Bahrami A., Soleimani A., Saeedi N., Rahmani F., Khazaei M. et al. The diagnostic, prognostic and therapeutic potential of circulating microRNAs in ovarian cancer.Int. J. Biochem. Cell Biol. 2020; 124: 105765. https://dx.doi.org/10.1016/j.biocel.2020.105765.
  14. Love M.I., Huber W., Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15(12): 550. https://dx.doi.org/10.1186/s13059-014-0550-8.
  15. Licursi V., Conte F, Fiscon G., Paci P. MIENTURNET: An interactive web tool for microRNA-target enrichment and network-based analysis. BMC Bioinformatics. 2019; 20(1):545. https://dx.doi.org/10.1186/s12859-019-3105-x.
  16. Glaab E., Baudot A., Krasnogor N., Schneider R., Valencia A. EnrichNet: network-based gene set enrichment analysis. Bioinformatics. 2012; 28(18): i451 7. https://dx.doi.org/10.1093/bioinformatics/bts389
  17. Pegtel D.M., Gould S.J. Exosomes. Annu. Rev. Biochem. 2019; 88: 487-514. https://dx.doi.org/10.1146/annurev-biochem-013118-111902.
  18. Kim K.M., Abdelmohsen K., Mustapic M., Kapogiannis D., Gorospe M. RNA in extracellular vesicles. Wiley Interdiscip. Rev. RNA. 2018; 8(4): 10.1002/ wrna.1413. https://dx.doi.org/10.1002/wrna.1413.
  19. Cho S., Mutlu L., Grechukhina O., Taylor H.C. Circulating microRNAs as potential biomarkers for endometriosis. Fertil. Steril. 2015; 103(5): 1252-60. e1. 103(5):1252- 103(5):1252-60. e1. https://dx.doi.org/10.1016/j.fertnstert.2015.02.013.
  20. Zafari N., Tarafdari A.M., Izadi P. A panel of plasma miRNAs 199b-3p, 224-5p and Let-7d-3p as non-invasive diagnostic biomarkers for endometriosis. Reprod. Sci. 2021; 28(4): 991-9. https://dx.doi.org/10.1007/s43032-020-00415-z.
  21. Vanhie A., O. D., Peterse D., Beckers A., Cuellar A., Fassbender A. et al. Plasma miRNAs as biomarkers for endometriosis. Hum. Reprod. 2019; 34(9): 1650-60. https://dx.doi.org/10.1093/humrep/dez116.
  22. Moustafa S., Burn M., Mamillapalli R., Nematian S., Flores V., Taylor H.S. Accurate diagnosis of endometriosis using serum microRNAs. Am. J. Obstet. Gynecol. 2020; 223(4): 557.e1-557.e11. https://dx.doi.org/10.1016/j.ajog.2020.02.050.
  23. Masoumi-Dehghi S., Babashah S. microRNA-141-3p-containing small extracellular vesicles derived from epithelial ovarian cancer cells promote endothelial cell angiogenesis through activating the JAK / STAT3 and NF-k B. signaling pathways. J. Cell Commun. Signal. 2020; 14(2): 233-44. https://dx.doi.org/10.1007/s12079-020-00548-5.
  24. Leonova A., Turpin V.E., Agarwal S.K., Leonardi M., Foster W.G. A critical appraisal of the circulating levels of differentially expressed microRNA in endometriosis. Biol. Reprod. 2021; 105(5): 1075-85. https://dx.doi.org/10.1093/biolre/ioab134.
  25. Truong G., Guanzon D., Kinhal V., Elfeky O., Lai A., Longo S. et al. Oxygen tension regulates the miRNA profile and bioactivity of exosomes released from extravillous trophoblast cells - Liquid biopsies for monitoring complications of pregnancy. PLoS One. 2017; 12(3): e0174514. https://dx.doi.org/10.1371/journal.pone.0174514.
  26. Hu J., Tang T., Zeng Z., Wu J., Tan X., Yan J. The expression of small RNAs in exosomes of follicular fluid altered in human polycystic ovarian syndrome. Peer J. 2020; 8: e8640. https://dx.doi.org/10.7717/peerj.8640.
  27. Papari E., Noruzinia M., Kashani L., Foster W.G. Identification of candidate microRNA markers of endometriosis with the use of next-generation sequencing and quantitative real-time polymerase chain reaction. Fertil. Steril. 2020; 113(6): 1232-41. https://dx.doi.org/10.1016/j.fertnstert.2020.01.026.
  28. Bjorkman S., Taylor H.S. MicroRNAs in endometriosis: biological function and emerging biomarker candidates. Biol. Reprod. 2019; 100(5): 1135-46. https://dx.doi.org/10.1093/biolre/ioz014.
  29. Roman-Canal B., Moiola C.P., Gatius S., Bonnin S., Ruiz-Miro M., Gonzalez E. et al. EV-associated miRNAs from peritoneal lavage are a source of biomarkers in endometrial cancer. Cancers (Basel). 2019; 11(6): 839. https://dx.doi.org/10.3390/cancers11060839.
  30. Taylor D.D., Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008; 110(1): 13-21. https://dx.doi.org/10.1016/j.ygyno.2008.04.033.
  31. Chen Y., Wang K., Xu Y., Guo P., Hong B., Cao Y. et al. Alteration of myeloid-derived suppressor cells, chronic inflammatory cytokines, and exosomal miRNA contribute to the peritoneal immune disorder of patients with endometriosis. Reprod. Sci. 2018; 26(8): 1130-8. https://dx.doi.org/10.1177/1933719118808923.
  32. Nakamura N., Terai Y., Nunode M., Kokunai K., Konishi H., Taga S. et al. The differential expression of miRNAs between ovarian endometrioma and endometriosis-associated ovarian cancer. J. Ovarian Res. 2020; 13(1): 51. https://dx.doi.org/10.1186/s13048-020-00652-5.
  33. Bjornetro T., Redalen K.R., Meltzer S., Thusyanthan N.S., Samiappan R., Jegerschold C. et al. An experimental strategy unveiling exosomal microRNAs 486-5p, 181a-5p and 30d-5p from hypoxic tumour cells as circulating indicators of high-risk rectal cancer. J. Extracell. Vesicles. 2019; 8(1): 1567219. https://dx.doi.org/10.1080/20013078.2019.1567219.
  34. Liu C., Li M., Hu Y., Shi N., Yu H., Liu H., Lian H. miR-486-5p attenuates tumor growth and lymphangiogenesis by targeting neuropilin-2 in colorectal carcinoma. Onco Targets Ther. 2016; 9: 2865-71. https://dx.doi.org/10.2147/ OTT.S103460.
  35. Narasimhan A., Ghosh S., Stretch C., Greiner R., Bathe O.F., Baracos V., Damaraju S. Small RNAome pro fi ling from human skeletal muscle: novel miRNAs and their targets associated with cancer cachexia. J. Cachexia Sarcopenia Muscle. 2017; 8(3): 405-16. https://dx.doi.org/10.1002/jcsm.12168.
  36. Liu J., Yoo J., Ho J.Y., Jung Y., Lee S., Hur S.Y., Choi Y.J. Plasma-derived exosomal miR-4732-5p is a promising noninvasive diagnostic biomarker for epithelial ovarian cancer. J. Ovarian Res. 2021; 14(1): 59. https://dx.doi.org/10.1186/s13048-021-00814-z.
  37. Cochrane D.R., Howe E.N., Spoelstra N.S., Richer J.K. Loss of miR-200c: a marker of aggressiveness and chemoresistance in female reproductive cancers. J. Oncol. 2010; 2010: 821717. https://dx.doi.org/10.1155/2010/821717.
  38. Cao Q., Lu K., Dai S., Hu Y., Fan W. Clinicopathological and prognostic implications of the miR-200 family in patients with epithelial ovarian cancer. Int. J. Clin. Exp. Pathol. 2014; 7(5): 2392-401.
  39. Gajek A., Gralewska P., Marczak A., Rogalska A. Current implications of microRNAs in genome stability and stress responses of ovarian cancer. Cancers (Basel). 2021; 13(11): 2690. https://dx.doi.org/10.3390/cancers13112690.
  40. Бобров М.Ю., Балашов И.С., Филиппова Е.С., Альмова И.К., Тимофеева А.В., Гусар В.А., Боровиков П.И., Хилькевич Е.Г., Чупрынин В.Д., Павлович С.В. Оценка экспрессии микроРНК в очагах ретроцервикаль-ного эндометриоза. Акушерство и гинекология. 2018; 6: 55-61. [Bobrov M.Yu., Balashov I.S., Filippova E.S., Almova I.K., Timofeeva A.V., Gusar V.A., Borovikov P.I., Khilkevich E.G., Chuprynin V.D., Pavlovich S.V. Assessment of microRNA expression in retrocervical endometriotic lesions. Akusherstvo i Ginekologiya/Obstetrics and Gynecology. 2018; 6: 55-61. (in Russian)]. https://dx.doi.org/10.18565/aig.2018.6.55-61.
  41. Бобров М.Ю., Балашов И.С., Филиппова Е.С., Альмова И.К., Хилькевич Е.Г., Павлович С.В., Наумов В.А., Боровиков П.И., Сухих Г.Т. Использование транскриптомных баз данных для анализа патогенетических факторов эндометриоза. Акушерство и гинекология. 2017; 4: 34-44. [Bobrov M.Yu., Balashov I.S., Filippova E.S., Almova I.K., Khilkevich E.G., Pavlovich S.V., Naumov V.A., Borovikov P.I., Sukhikh G.T. Use of transcriptomic databases for the analysis of pathogenetic factors of endometriosis. Akusherstvo i Ginekologiya/Obstetrics and Gynecology. 2017; 4: 34-44. (in Russian)]. https://dx.doi.org/10.18565/aig.2017.4.34-44.

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2022 Bionika Media

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies