Open access peer-reviewed chapter

microRNA and Overcoming the Challenges of Their Use in the Diagnosis of Endometriosis

Written By

Victoria Turpin, Anna Leonova, Sanjay K. Agarwal and Warren G. Foster

Submitted: 30 December 2019 Reviewed: 22 January 2020 Published: 19 February 2020

DOI: 10.5772/intechopen.91324

From the Edited Volume

Endometriosis

Edited by Courtney Marsh

Chapter metrics overview

848 Chapter Downloads

View Full Metrics

Abstract

Endometriosis is a common estrogen dependent and inflammatory disease affecting approximately 176 million women worldwide. Currently, the time between onset of symptoms and a definitive diagnosis has been reported by several international studies to range from 6 to 12 years. Presently, laparoscopic surgery followed by histopathological confirmation of lesions remains the gold standard for diagnosis. In part because of cost and invasiveness, current trends favor reduced laparoscopic surgeries in preference of the non-surgical diagnosis of endometriosis. However, the search for a clinical marker or markers of endometriosis that provide equal or similar sensitivity and specificity to laparoscopy has remained elusive. Thus, the search for a diagnostic test for the diagnosis of endometriosis continues to be a high priority research and clinical issue. Recent studies have reported favorable results with microRNA; however, lack of replication and absence of validation suggest that circulating miRNA may not be reliable for clinical use. Use of different screening platforms together with divergent methods may account for some of the lack or reproducibility in the literature. Herein we critically assess the recent literature and explore sources for discrepant findings. We suggest that prospective studies using validated reference miRNA to normalize results together with improved study design may yet reveal a suitable diagnostic marker or panel of markers for the diagnosis of endometriosis.

Keywords

  • microRNA
  • miRNA
  • diagnosis
  • plasma
  • endometriosis

1. Background

Endometriosis is a common estrogen dependent and progesterone resistant disease of unknown cause characterized by growth of endometrial cells outside the uterine cavity [1]. It is estimated that 6–11% of all women are affected by endometriosis reaching an estimated 176 million women globally [2]. A chronic painful disease [3], endometriosis causes substantial health distress and interference with normal activities including work resulting in an average loss of 10.8 h/week from work [2] all leading to diminished quality of life (QOL) for affected women and their families. Chronic pelvic pain and infertility are common symptoms of endometriosis that bring women with this disease to seek medical attention. Approximately 71–87% of all women experiencing chronic pelvic pain and 50% of infertile women are diagnosed with endometriosis [4]. Thus, women with endometriosis report significant health distress and interference with normal activities including work and leisure time activities all leading to a deleterious effect upon women’s social functioning, emotional well-being, employment, and vitality [5].

An important obstacle to the timely diagnosis and effective management of endometriosis is the lack of a simple diagnostic test. Diagnosis has been reported to be delayed by between 6 and 12 years with an average of 9 years from the onset of symptoms to definitive diagnosis [6]. Hence, identification of a clinical tool for the diagnosis of endometriosis has become a high priority research objective [2, 7, 8]. Health care providers and patients face a number of challenges in arriving at a diagnosis of endometriosis including: early age at onset of symptoms, normalization of pain by primary care providers, and suppression of symptoms through intermittent use of oral contraceptive pills [9]. Symptoms of endometriosis are shared with many other diseases including autoimmune diseases, cancer, irritable bowel syndrome (IBS), and musculoskeletal abnormalities. Therefore, an ideal biomarker of endometriosis must differentiate between endometriosis and other explanations for patient symptoms. In addition, clinical markers should be as minimally invasive as possible, affordable and convenient to use with the added benefit of providing insight into potential treatment response. Furthermore, the ideal biomarker must provide equivalent or similar outcome measures of sensitivity, specificity, positive, and negative predictive values to laparoscopy.

Currently, the gold standard for diagnosis remains visualization of endometriotic lesions typically by laparoscopy followed by histopathological confirmation of disease [10]. Current trends favoring the non-surgical diagnosis of endometriosis increase the pressure to identify novel clinical markers of endometriosis. Despite a plethora of biochemical differences in the peripheral circulation, peritoneal fluid, and endometrial tissues of women with endometriosis compared to healthy controls, no marker has been found with adequate sensitivity or specificity to challenge laparoscopy for the diagnosis of endometriosis whether used alone or in a panel of clinical markers [11, 12, 13, 14, 15, 16]. However, reports of promising results have been brought forward in the literature from which epigenetic markers are potentially the most exciting.

Advertisement

2. Candidate clinical markers of endometriosis

Multiple gene and protein expression levels have been documented in women with endometriosis compared to controls; however, none have yielded reliable clinical markers of disease. Recent studies investigating the mechanisms controlling gene expression have produced promising results. Several histone modifications have been associated with endometriosis. For example, endometriotic stromal cells (ESC) have a lower global acetylation level of H3, and histone deacetylases 1 and 2 (HDAC1 and HDAC2) were upregulated compared to women without endometriosis [17]. Furthermore, histone deacetylase inhibitor (HDACI) treatment promoted apoptosis by reactivating the silenced chromatin [18]. G-protein-coupled estrogen receptor (GPER) expression and proliferation of endometriotic cells was inhibited by treatment with the HDACI’s romidepsin and vorinostat [19]. These data suggest that histone modifications are involved in the pathophysiology of endometriosis and that HDACI’s are promising agents for endometriosis treatment. However, use of histone markers in the diagnosis of endometriosis has yet to be explored.

Long-chain non-coding RNA (lnc-RNAs) are 200–100,000 bp RNA molecules which do not encode for protein, but are involved in transcriptional and post-transcriptional regulation of gene expression [20]. They are the most common non-coding RNAs and are involved in cell proliferation, differentiation, and apoptosis; all processes central in the pathobiology of endometriosis [21]. Some lnc-RNAs proposed as diagnostic markers of endometriosis include: H19 [22], CHL1-AS2 [23, 24], AC002454.1 [25], lncRNA SRA (steroid receptor RNA activator) [26], MALAT-1 [27], and LINC01279 [28]. Results of a recent study revealed the lnc-RNA are carried in circulating extracellular vesicles in women with endometriosis [29]. However, use of lnc-RNAs in the diagnosis of endometriosis has not been evaluated in a prospective study of women with symptoms suggestive of endometriosis with an independent validation step and thus their clinical utility remains uncertain.

Several recent studies have documented aberrant expression of multiple microRNAs (miRNAs) in the eutopic and ectopic endometrium of women with endometriosis [30, 31, 32, 33, 34, 35, 36, 37]. miRNAs are short non-coding RNAs, 19–25 nucleotides long, that negatively regulate mRNA translation by repressing the protein translational machinery or degrading their target transcripts. Greater than 2000 mature human miRNA sequences have been identified and are thought to regulate approximately 50% of all protein coding genes. Multiple recent studies have documented differential microRNA (miRNA) expression in endometriotic tissues compared to eutopic endometrium of women with endometriosis and controls [33, 38, 39, 40]. miRNA are thought to hold promise as diagnostic biomarkers of disease because they are post-transcriptional regulators of gene expression that are stably expressed over time in bodily fluids and tissues [41]. Briefly, miRNA regulate protein expression through binding to and inhibiting the translation of mRNA transcripts into protein [42]. miRNAs are synthesized in the cytoplasm from nucleic hairpin intermediates (pre-miRNA) [43] which are then processed to yield mature miRNA that resist RNase degradation [41]. miRNA form an RNA-induced silencing complex (RISC) with Argonaute, Dicer, TAR RNA binding protein (TRBP) and protein activator of PKR (PACT) to post-transcriptionally regulate genes by binding to the 3′ region of the mRNA transcript and inhibiting translation [44].

In the early 2000s, several studies proposed that circulating levels of miRNA are differentially expressed in women with endometriosis compared to controls and thus could have diagnostic value [30, 31, 45]. Different methods including in situ hybridization, targeted RT-PCR and several different screening platforms including miRNA based microarrays, next generation sequencing and bio-informatics followed by RT-PCR validation have subsequently revealed a broad spectrum of miRNAs that are differentially expressed in women with endometriosis compared to control groups [29, 30, 31, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56]. However, to date, only the results for hsa-miR-451a [47, 48], 199a-5p [31, 54] and hsa-miR-141-3p [31, 49] have been successfully replicated in more than one study (Table 1). For the vast majority of miRNAs, differential expression has only been reported in a single study or the results for a few miRNAs have not been replicated by other investigators. For example, circulating levels of hsa-miR-145 were lower in women with endometriosis compared to controls [31] whereas hsa-miR-145 levels did not differ [47] or were higher in women with endometriosis compared to the control groups [50]. We postulate that divergent results may be the consequence of different screening platforms and technologies used to identify candidate miRNA markers of disease [57, 58, 59] and control group characteristics. Moreover, we suggest that different reference material used to quantify circulating miRNA levels are an additional source of variation.

Fluid Case/control (N) Cases Controls Reference miRNA Cycle stage Differentially expressed hsa-miR’s Citation
Serum 60/25 Stages I–IV Symptomatic RNU6 NS 199a and 122, ↓9 * , 145 * , 141 * , and 542-3p [31]
Serum 24/24 Stage III and IV Symptomatic RNU6 P vs. S ↓let-7b, c, d, e, f (P) and 135a (S) [46]
Serum 24/24 Stage III and IV Symptomatic RNU6 NS ↓3613-5p and 6755-3p ↑18a-5p, 125b-5p, 143-3p, 150-5p 342-3p, 451a and 500a-3p [47]
Serum 41/40 Stages I–IV Symptomatic (n = 20) and Healthy (n = 20) RNU6 NS 451a [48]
Serum 30/20 Stages I–II Infertile cel-miR-39 ND ↓30c-5p, 127-3p, 99b-5p, 15b-5p, 20a-5p, and ↑424-3p, 185-5p [52]
Serum 45/35 Stages I–IV Symptomatic RNU6 ND 122 and 199a [54]
Serum 40/25 Stages I–IV Healthy 18 s mRNA ND ↓199a-5p [55]
Plasma 23/23 Stage III and IV Symptomatic miR-16 ND ↓17-5p, 20a and 22 [30]
Plasma 61/30 Stages I–IV Symptomatic (n = 35) and Healthy (n = 30) miR-30e and 99a NS ↓200a-3p and 141-3p [49]
Plasma 55/23 Stages I–IV Symptomatic miR-103-3p ND 145 (stages I and II), ↓31(stages I–IV) [50]
Plasma Variable Stage I–IV Symptomatic (n = 8–39) Healthy (n = 8) RNU6 vs. miR-16 vs. miR-30b NS ↓139-3p, 155 and 574-3p [51]
Plasma 51/41 Stages I–IV Symptomatic miR-106a-5p, 199a-3p, 150-5p, 425-5p, 125a-5p, and 30e-5p NS ↓miR154-5p and 378a-3p, ↑196b-5p and 33a-5p [53]
Plasma 60/30 Stages I–IV Symptomatic miR-28-3p and 423-3p S * 125b-5p, 28-5p, 29a-3p [56]
Plasma 33/20 NR Healthy miR-132 NR ↑15b, 16, 191, 195, 1973, 1979, and 4284 [45]
Plasma 6/4 Stages III–IV Healthy RNU6 NR ↓375, 27a-3p, and 30d-5p [29]

Table 1.

Summary of miRNA’s differentially expressed by microarray and RT-PCR in women with endometriosis (cases) compared to controls.

Direction could not be ascertained from the published report.


miRNA in bold have been replicated by at least one other group of investigators.

S = significant effect of menstrual cycle stage, NS = not significant, ND = not determined, NR = not reported, differential miR expression was either increased (↑) or decreased (↓) in women with endometriosis compared to controls.

While RNU6 has been widely used in the general miRNA literature to normalize miRNA expression in tissue, abundance and stability of expression have not been evaluated for circulating miRNA expression in women with endometriosis. Furthermore, RNU6 has low stability and abundance that is greatly influenced by sample storage and processing and the Cp values of RNU6 are highly variable from miRNA Cp values [51, 60, 61]. Similarly, the abundance and stability of miR-16-5p levels in the serum of women with endometriosis is uncertain but variable from the Cp values of miRNA targets [51]. Furthermore, circulating levels of miR-16-5p are altered by inflammation and stress [62, 63] and thus we suggest that both RNU6 and miR-16-5p are not suitable for normalization of circulating miRNA levels in women with endometriosis.

Advertisement

3. Effect of reference miRNA used to normalize results

While serum RNU6 has been widely used as the reference miRNA in prior endometriosis studies [29, 31, 46, 47, 48, 54], its levels have previously been reported to be unstable, unreliable, and a poor reference for miRNA since it is not processed or protected in the same way as miRNA [61, 63]. Therefore, we suggest that choice of reference miRNA can influence ability to detect significant differences and the direction of significant differences elicited. Previous studies report that hsa-miR-451a is upregulated in women with endometriosis compared to symptomatic controls [47] and compared to both symptomatic and asymptomatic (healthy) control groups [48]. Both prior studies employed RNU6 as a reference. While hsa-miR-451a has been found to act as a tumor suppressor [64, 65], it is also a marker of hemolysis [66] and thus we suggest that care should be employed to exclude samples with hemolysis before analysis. The miRNA ratio of hsa-miR-451a and hsa-miR-23a-3p has been employed by others [56, 67] to monitor for sample hemolysis. Therefore, we suggest that hsa-miR-451a has limited value as a candidate marker of endometriosis.

Advertisement

4. Effect of control group definition

Several studies have employed healthy women as their control population [29, 45, 48, 49, 51, 55], thus allowing circulating miRNA levels in women with endometriosis to be compared to symptomatic and asymptomatic healthy control populations. While the majority of previous reports employed symptomatic controls [30, 31, 46, 47, 48, 49, 50, 51, 53, 54, 56], hsa-miR-16-5p [30, 51] RNU6 (the most common) reference material used to normalize miRNA expression [31, 46, 47, 48, 51, 54]; reference materials that are unsuitable for normalizing serum miRNA expression. In our experience, differential miRNA expression was dependent upon whether comparisons were made with asymptomatic compared to symptomatic controls. Therefore, we suggest that control group characteristics on the differential expression of candidate miRNA in women with endometriosis merits further investigation.

While, lack of replication, absence of validation of results, and poor sensitivity and specificity currently limit the value of miRNA as diagnostic markers of endometriosis [51], we propose that usefulness of miRNA for the diagnosis of endometriosis cannot be evaluated without appropriate determination of appropriate reference miRNA.

Advertisement

5. Future directions

Although identification of clinical markers of endometriosis has long been sought, none has so far been suitable to displace laparoscopy as the gold standard for diagnosis. Endometriosis is a complex heterogenous disease with variable presentation whose symptoms are easily confused with other clinical problems. Since endometriosis is detectable with high frequency amongst asymptomatic women [68] surgical exclusion of disease in the control group is essential to prevent biasing results towards the null. Consequently, we suggest that control or reference group definition is important. Numerous prior studies reporting differential miRNA expression in women with endometriosis have employed asymptomatic women as their reference population [29, 45, 48, 49, 51, 55]. However, healthy women without symptoms of endometriosis and without evidence of endometriosis by laparoscopy (asymptomatic control) and symptomatic women without evidence of disease at the time of laparoscopy (symptomatic control) are functionally different, yet both groups continue to be employed as controls in contemporary studies. Results from our laboratory suggest that inclusion of asymptomatic controls can produce misleading results and thus speculate that restricting the control group to symptomatic controls in future studies may improve reproducibility of results. In addition to control group, we propose that the use of validated reference miRNA to normalize results also affects detection of levels of miRNA differentially expressed in women with endometriosis compared to controls.

Having identified candidate miRNA for the diagnosis of endometriosis it will be important to determine their relationship with pelvic pain as well as response to treatment. In the absence of this data the potential prognostic value of candidate markers of endometriosis remains uncertain. We also propose that future studies with robust sample size will be needed to clarify the relationship between circulating miRNA levels and menstrual cycle phase. Studies reporting menstrual cycle stage and circulating miRNA levels are thus far have produced equivocal results [31, 46, 47, 48, 49, 51, 53, 56]. Furthermore, lesion type (endometrioma, peritoneal endometriosis, deep infiltrating endometriosis) are biologically distinct and thus a single clinical marker is unlikely to be dysregulated in all lesion types and thus a panel of markers may be more relevant. Furthermore, duration of disease and age of lesion may also present with functional differences. Therefore, discovery of clinical markers should describe the lesion types present in study participants. The influence of study participant age and body mass index are also important variables associated with pelvic pain and disease severity that are frequently not considered in analyses of clinical markers of endometriosis. Finally, the functional role of candidate markers in endometriosis has the potential to suggest therapeutic targets for additional research.

Advertisement

6. Summary and conclusions

Use of reference miRNA that may not be ideal for normalization of results may account for noted weaknesses in the literature. Use of validated reference miRNA markers and careful control of sample condition for potential confounders should improve study replication. Finally, although circulating miRNA levels have low variability in women with endometriosis, it will be necessary for discovery phases to include a large number of study participants to control for participant age, menstrual cycle stage, BMI, stage of disease, and type of lesions. Thus, we suggest that despite set-backs with reproducibility of results, it may be too soon to judge the diagnostic potential of miRNA.

Advertisement

Acknowledgments

Funding support for this project was provided by a research operating grant (MOP142230) from the Canadian Institutes of Health Research to WGF. The authors are grateful to Ms. Annette Bullen for her assistance in study participant recruitment, access to clinical data, and sample collection. The assistance of the Endo@Mac surgical team (Drs. Nick Leyland, Sarah Scatollon, and Dustin Costescu) with access to study participants, staging of disease, and collection of blood samples is greatly appreciated.

References

  1. 1. Sampson J. Peritoneal endometriosis due to the menstrual dissemination of endometrial tissue into the peritoneal cavity. American Journal of Obstetrics and Gynecology. 1927;14:422-469
  2. 2. Rogers PA, Adamson GD, Al-Jefout M, Becker CM, D’Hooghe TM, Dunselman GA, et al. Research priorities for endometriosis. Reproductive Sciences. 2017;24(2):202-226
  3. 3. Kennedy S, Bergqvist A, Chapron C, D'Hooghe T, Dunselman G, Greb R, et al. ESHRE guideline for the diagnosis and treatment of endometriosis. Human Reproduction. 2005;20(10):2698-2704
  4. 4. Aredo JV, Heyrana KJ, Karp BI, Shah JP, Stratton P. Relating chronic pelvic pain and endometriosis to signs of sensitization and myofascial pain and dysfunction. Seminars in Reproductive Medicine. 2017;35(1):88-97
  5. 5. Jones G, Jenkinson C, Kennedy S. The impact of endometriosis upon quality of life: A qualitative analysis. Journal of Psychosomatic Obstetrics & Gynecology. 2004;25(2):123-133
  6. 6. Agarwal SK, Chapron C, Giudice LC, Laufer MR, Leyland N, Missmer SA, et al. Clinical diagnosis of endometriosis: A call to action. American Journal of Obstetrics and Gynecology. 2019;220(4):354.e351-354.e312
  7. 7. Rogers PA, D’Hooghe TM, Fazleabas A, Giudice LC, Montgomery GW, Petraglia F, et al. Defining future directions for endometriosis research: Workshop report from the 2011 world congress of endometriosis in Montpellier, France. Reproductive Sciences. 2013;20(5):483-499
  8. 8. Rogers PA, D’Hooghe TM, Fazleabas A, Gargett CE, Giudice LC, Montgomery GW, et al. Priorities for endometriosis research: Recommendations from an international consensus workshop. Reproductive Sciences. 2009;16(4):335-346
  9. 9. Ballard K, Lowton K, Wright J. What’s the delay? A qualitative study of women's experiences of reaching a diagnosis of endometriosis. Fertility and Sterility. 2006;86(5):1296-1301
  10. 10. Wykes CB, Clark TJ, Khan KS. Accuracy of laparoscopy in the diagnosis of endometriosis: A systematic quantitative review. BJOG. 2004;111(11):1204-1212
  11. 11. May KE, Conduit-Hulbert SA, Villar J, Kirtley S, Kennedy SH, Becker CM. Peripheral biomarkers of endometriosis: A systematic review. Human Reproduction Update. 2010;16(6):651-674
  12. 12. May KE, Villar J, Kirtley S, Kennedy SH, Becker CM. Endometrial alterations in endometriosis: A systematic review of putative biomarkers. Human Reproduction Update. 2011;17(5):637-653
  13. 13. Fassbender A, Burney RO, O DF, D'Hooghe T, Giudice L. Update on biomarkers for the detection of endometriosis. BioMed Research International. 2015;2015:130854
  14. 14. Fassbender A, Vodolazkaia A, Saunders P, Lebovic D, Waelkens E, De MB, et al. Biomarkers of endometriosis. Fertility and Sterility. 2013;99(4):1135-1145
  15. 15. Toor K, Wessels JM, Agarwal SK, Leyland N, Foster WG. Clinical markers of endometriosis: Have we been too quick to judge? Medical Hypotheses. 2014;82(4):493-501
  16. 16. Nisenblat V, Bossuyt PM, Shaikh R, Farquhar C, Jordan V, Scheffers CS, et al. Blood biomarkers for the non-invasive diagnosis of endometriosis. Cochrane Database of Systematic Reviews. 2016;5:CD012179
  17. 17. Monteiro JB, Colón-Díaz M, García M, Gutierrez S, Colón M, Seto E, et al. Endometriosis is characterized by a distinct pattern of histone 3 and histone 4 lysine modifications. Reproductive Sciences. 2014;21(3):305-318
  18. 18. Cameron EE, Bachman KE, Myöhänen S, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nature Genetics. 1999;21(1):103-107
  19. 19. Imesch P, Samartzis EP, Dedes KJ, Fink D, Fedier A. Histone deacetylase inhibitors down-regulate G-protein-coupled estrogen receptor and the GPER-antagonist G-15 inhibits proliferation in endometriotic cells. Fertility and Sterility. 2013;100(3):770-776
  20. 20. Spizzo R, Almeida MI, Colombatti A, Calin GA. Long non-coding RNAs and cancer: A new frontier of translational research? Oncogene. 2012;31(43):4577-4587
  21. 21. Johnsson P, Lipovich L, Grandér D, Morris KV. Evolutionary conservation of long non-coding RNAs; sequence, structure, function. Biochimica et Biophysica Acta. 2014;1840(3):1063-1071
  22. 22. Ghazal S, McKinnon B, Zhou J, Mueller M, Men Y, Yang L, et al. H19 lncRNA alters stromal cell growth via IGF signaling in the endometrium of women with endometriosis. EMBO Molecular Medicine. 2015;7(8):996-1003
  23. 23. Sun PR, Jia SZ, Lin H, Leng JH, Lang JH. Genome-wide profiling of long noncoding ribonucleic acid expression patterns in ovarian endometriosis by microarray. Fertility and Sterility. 2014;101(4):1038-1046.e1037
  24. 24. Zhang C, Wu W, Ye X, Ma R, Luo J, Zhu H, et al. Aberrant expression of CHL1 gene and long non-coding RNA CHL1-AS1, CHL1-AS2 in ovarian endometriosis. European Journal of Obstetrics, Gynecology, and Reproductive Biology. 2019;236:177-182
  25. 25. Wang Y, Li Y, Yang Z, Liu K, Wang D. Genome-wide microarray analysis of long non-coding RNAs in Eutopic secretory endometrium with endometriosis. Cellular Physiology and Biochemistry. 2015;37(6):2231-2245
  26. 26. Lin K, Zhan H, Ma J, Xu K, Wu R, Zhou C, et al. Silencing of SRA1 regulates ER expression and attenuates the growth of stromal cells in ovarian endometriosis. Reproductive Sciences. 2017;24(6):836-843
  27. 27. Liu H, Zhang Z, Xiong W, Zhang L, Du Y, Liu Y, et al. Long non-coding RNA MALAT1 mediates hypoxia-induced pro-survival autophagy of endometrial stromal cells in endometriosis. Journal of Cellular and Molecular Medicine. 2019;23(1):439-452
  28. 28. Liu J, Wang Q , Zhang R, Zhang C, Lin J, Huang X. Identification of LINC01279 as a cell cycle-associated long non-coding RNA in endometriosis with GBA analysis. Molecular Medicine Reports. 2018;18(4):3850-3858
  29. 29. Khalaj K, Miller JE, Lingegowda H, Fazleabas AT, Young SL, Lessey BA, et al. Extracellular vesicles from endometriosis patients are characterized by a unique miRNA-lncRNA signature. JCI Insight. 2019;4(18):e12886
  30. 30. Jia SZ, Yang Y, Lang J, Sun P, Leng J. Plasma miR-17-5p, miR-20a and miR-22 are down-regulated in women with endometriosis. Human Reproduction. 2013;28(2):322-330
  31. 31. Wang WT, Zhao YN, Han BW, Hong SJ, Chen YQ . Circulating microRNAs identified in a genome-wide serum microRNA expression analysis as noninvasive biomarkers for endometriosis. Journal of Clinical Endocrinology and Metabolism. 2013;98(1):281-289
  32. 32. Liu S, Gao S, Wang XY, Wang DB. Expression of miR-126 and Crk in endometriosis: miR-126 may affect the progression of endometriosis by regulating Crk expression. Archives of Gynecology and Obstetrics. 2012;285(4):1065-1072
  33. 33. Shen L, Yang S, Huang W, Xu W, Wang Q , Song Y, et al. MicroRNA23a and microRNA23b deregulation derepresses SF-1 and upregulates estrogen signaling in ovarian endometriosis. Journal of Clinical Endocrinology and Metabolism. 2013;98(4):1575-1582
  34. 34. Ramon LA, Braza-Boils A, Gilabert-Estelles J, Gilabert J, Espana F, Chirivella M, et al. microRNAs expression in endometriosis and their relation to angiogenic factors. Human Reproduction. 2011;26(5):1082-1090
  35. 35. Gilabert-Estelles J, Braza-Boils A, Ramon LA, Zorio E, Medina P, Espana F, et al. Role of microRNAs in gynecological pathology. Current Medicinal Chemistry. 2012;19(15):2406-2413
  36. 36. Braza-Boils A, Mari-Alexandre J, Gilabert J, Sanchez-Izquierdo D, Espana F, Estelles A, et al. MicroRNA expression profile in endometriosis: Its relation to angiogenesis and fibrinolytic factors. Human Reproduction. 2014;29(5):978-988
  37. 37. Braza-Boils A, Gilabert-Estelles J, Ramon LA, Gilabert J, Mari-Alexandre J, Chirivella M, et al. Peritoneal fluid reduces angiogenesis-related microRNA expression in cell cultures of endometrial and endometriotic tissues from women with endometriosis. PLoS ONE. 2013;8(4):e62370
  38. 38. Haikalis ME, Wessels JM, Leyland NA, Agarwal SK, Foster WG. miRNA expression pattern differs depending on endometriosis lesion type. Biology of Reproduction. 2018;98(5):623-633
  39. 39. Burney RO, Hamilton AE, Aghajanova L, Vo KC, Nezhat CN, Lessey BA, et al. MicroRNA expression profiling of eutopic secretory endometrium in women with versus without endometriosis. Molecular Human Reproduction. 2009;15(10):625-631
  40. 40. Laudanski P, Charkiewicz R, Tolwinska A, Szamatowicz J, Charkiewicz A, Niklinski J. Profiling of selected MicroRNAs in proliferative Eutopic endometrium of women with ovarian endometriosis. BioMed Research International. 2015;2015:760698
  41. 41. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Research. 2008;18(10):997-1006
  42. 42. Bartel DP. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281-297
  43. 43. Lee Y, Jeon K, Lee JT, Kim S, Kim VN. MicroRNA maturation: Stepwise processing and subcellular localization. The EMBO Journal. 2002;21(17):4663-4670
  44. 44. Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K, et al. TRBP recruits the dicer complex to Ago2 for microRNA processing and gene silencing. Nature. 2005;436(7051):740-744
  45. 45. Suryawanshi S, Vlad AM, Lin HM, Mantia-Smaldone G, Laskey R, Lee M, et al. Plasma microRNAs as novel biomarkers for endometriosis and endometriosis-associated ovarian cancer. Clinical Cancer Research. 2013;19(5):1213-1224
  46. 46. Cho S, Mutlu L, Grechukhina O, Taylor HS. Circulating microRNAs as potential biomarkers for endometriosis. Fertility and Sterility. 2015;103(5):1252-1260 e1251
  47. 47. Cosar E, Mamillapalli R, Ersoy GS, Cho S, Seifer B, Taylor HS. Serum microRNAs as diagnostic markers of endometriosis: A comprehensive array-based analysis. Fertility and Sterility. 2016;106(2):402-409
  48. 48. Nothnick WB, Falcone T, Joshi N, Fazleabas AT, Graham A. Serum miR-451a levels are significantly elevated in women with endometriosis and recapitulated in baboons (Papio anubis) with experimentally-induced disease. Reproductive Sciences. 2017;24(8):1195-1202
  49. 49. Rekker K, Saare M, Roost AM, Kaart T, Soritsa D, Karro H, et al. Circulating miR-200-family micro-RNAs have altered plasma levels in patients with endometriosis and vary with blood collection time. Fertility and Sterility. 2015;104(4):938-946 e932
  50. 50. Bashti O, Noruzinia M, Garshasbi M, Abtahi M. miR-31 and miR-145 as potential non-invasive regulatory biomarkers in patients with endometriosis. Cell Journal. 2018;20(2):293
  51. 51. Nisenblat V, Sharkey DJ, Wang Z, Evans SF, Healey M, Ohlsson Teague EMC, et al. Plasma microRNAs display limited potential as diagnostic tools for endometriosis. The Journal of Clinical Endocrinology and Metabolism. 2019;104(6):1999-2022
  52. 52. Wang L, Huang W, Ren C, Zhao M, Jiang X, Fang X, et al. Analysis of serum microRNA profile by Solexa sequencing in women with endometriosis. Reproductive Sciences. 2016;23(10):1359-1370
  53. 53. Pateisky P, Pils D, Szabo L, Kuessel L, Husslein H, Schmitz A, et al. Hsa-miRNA-154-5p expression in plasma of endometriosis patients is a potential diagnostic marker for the disease. Reproductive Biomedicine Online. 2018;37(4):449-466
  54. 54. Maged AM, Deeb WS, El Amir A, Zaki SS, El Sawah H, Al Mohamady M, et al. Diagnostic accuracy of serum miR-122 and miR-199a in women with endometriosis. International Journal of Gynaecology and Obstetrics. 2018;141(1):14-19
  55. 55. Hsu CY, Hsieh TH, Tsai CF, Tsai HP, Chen HS, Chang Y, et al. miRNA-199a-5p regulates VEGFA in endometrial mesenchymal stem cells and contributes to the pathogenesis of endometriosis. The Journal of Pathology. 2014;232(3):330-343
  56. 56. Vanhie A, O D, Peterse D, Beckers A, Cuéllar A, Fassbender A, et al. Plasma miRNAs as biomarkers for endometriosis. Human Reproduction. 2019;34(9):1650-1660
  57. 57. Mestdagh P, Hartmann N, Baeriswyl L, Andreasen D, Bernard N, Chen C, et al. Evaluation of quantitative miRNA expression platforms in the microRNA quality control (miRQC) study. Nature Methods. 2014;11(8):809-815
  58. 58. Kok MGM, de Ronde MWJ, Moerland PD, Ruijter JM, Creemers EE, Pinto-Sietsma SJ. Small sample sizes in high-throughput miRNA screens: A common pitfall for the identification of miRNA biomarkers. Biomolecular Detection and Quantification. 2018;15:1-5
  59. 59. Pritchard CC, Cheng HH, Tewari M. MicroRNA profiling: Approaches and considerations. Nature Reviews. Genetics. 2012;13(5):358-369
  60. 60. Graham A, Falcone T, Nothnick WB. The expression of microRNA-451 in human endometriotic lesions is inversely related to that of macrophage migration inhibitory factor (MIF) and regulates MIF expression and modulation of epithelial cell survival. Human Reproduction. 2015;30(3):642-652
  61. 61. Xiang M, Zeng Y, Yang R, Xu H, Chen Z, Zhong J, et al. U6 is not a suitable endogenous control for the quantification of circulating microRNAs. Biochemical and Biophysical Research Communications. 2014;454(1):210-214
  62. 62. Wang H, Zhang P, Chen W, Feng D, Jia Y, Xie LX. Evidence for serum miR-15a and miR-16 levels as biomarkers that distinguish sepsis from systemic inflammatory response syndrome in human subjects. Clinical Chemistry and Laboratory Medicine. 2012;50(8):1423-1428
  63. 63. Katsuura S, Kuwano Y, Yamagishi N, Kurokawa K, Kajita K, Akaike Y, et al. MicroRNAs miR-144/144* and miR-16 in peripheral blood are potential biomarkers for naturalistic stress in healthy Japanese medical students. Neuroscience Letters. 2012;516(1):79-84
  64. 64. Liu G, Xu Z, Hao D. MicroRNA-451 inhibits neuroblastoma proliferation, invasion and migration by targeting macrophage migration inhibitory factor. Molecular Medicine Reports. 2016;13(3):2253-2260
  65. 65. Xu H, Mei Q , Shi L, Lu J, Zhao J, Fu Q . Tumor-suppressing effects of miR451 in human osteosarcoma. Cell Biochemistry and Biophysics. 2014;69(1):163-168
  66. 66. Shkurnikov MY, Knyazev EN, Fomicheva KA, Mikhailenko DS, Nyushko KM, Saribekyan EK, et al. Analysis of plasma microRNA associated with Hemolysis. Bulletin of Experimental Biology and Medicine. 2016;160(6):748-750
  67. 67. Blondal T, Jensby Nielsen S, Baker A, Andreasen D, Mouritzen P, Wrang Teilum M, et al. Assessing sample and miRNA profile quality in serum and plasma or other biofluids. Methods. 2013;59(1):S1-S6
  68. 68. Rawson JM. Prevalence of endometriosis in asymptomatic women. Journal of Reproductive Medicine. 1991;36(7):513-515

Written By

Victoria Turpin, Anna Leonova, Sanjay K. Agarwal and Warren G. Foster

Submitted: 30 December 2019 Reviewed: 22 January 2020 Published: 19 February 2020