Skip to main content

A promising future for endometriosis diagnosis and therapy: extracellular vesicles - a systematic review

Abstract

Endometriosis is a chronic, inflammatory gynaecological disease that can have severe negative impacts on quality of life and fertility, placing burden on patients and the healthcare system. Due to the heterogeneous nature of endometriosis, and the lack of correlation between symptom and surgical disease severity, diagnosis and treatment remain a significant clinical challenge. Extracellular vesicles (EVs) are biologically active particles containing molecular cargo involved in intercellular communication, that can be exploited for diagnostic and therapeutic purposes.

We systematically reviewed studies exploring EVs and their role in endometriosis, specifically addressing diagnostic and therapeutic potential and current understanding of pathophysiology. Five databases (Pubmed, Embase, Medline, Web of Science, Google Scholar) were searched for keywords ‘endometriosis’ and either ‘extracellular vesicles’ or ‘exosomes’.

There were 28 studies included in the review. Endometrium derived EVs contribute to the development of endometriosis. EVs derived from endometriosis lesions contribute to angiogenesis, immunomodulation and fibrosis. Such EVs can be detected in blood, with early data demonstrating utility in diagnosis and recurrence detection. EV isolation techniques varied between studies and only eight of twenty-eight studies fully characterised EVs according to current recommended standards. Reporting/type of endometriosis was limited across studies. Varied patient population, type of sample and isolation techniques created bias and difficulty in comparing studies.

EVs hold promise for improving care for symptomatic patients who have never had surgery, as well as those with recurrent symptoms after previous surgery. We encourage further EV research in endometriosis with the inclusion of rigorous reporting of both the patient population and technical methodology used, with the ultimate goal of achieving clinical utility for diagnosis, prognosis and eventually treatment.

Background

Endometriosis is a heterogeneous condition with wide variation in reported prevalence [1], estimated to affect 10% of women worldwide (190 million women globally) [2]. The condition is defined by the presence of endometrial-like glands and stroma in locations outside of the endometrial cavity; and can manifest with dysmenorrhoea, subfertility and a heterogeneous range of other symptoms with little correlation between disease and symptom severity [3].

The diagnosis of endometriosis at present is reliant on clinical history and examination, ultrasound, and ultimately laparoscopic surgery and biopsy [3, 4]. Whilst endometriomas and deep infiltrating endometriosis can be diagnosed with increasing reliability by ultrasound [3], the more common manifestation of superficial peritoneal disease is poorly predicted without surgery [5].

Around half of women who have clinically suspected endometriosis undergo surgery to confirm the diagnosis [6]. Clinical suspicion of endometriosis is an inconsistent predictor of disease; of women undergoing laparoscopy for suspected endometriosis, 18–77% have surgically confirmed disease [7], although recent evidence suggests that the rates of microscopic endometriosis in women with “negative laparoscopy” may be up to 39% [8]. There is a strong need for a reliable non-invasive diagnostic tool to reduce the need for laparoscopy to diagnose or exclude endometriosis.

Management of endometriosis is generally divided into conservative, hormonal and surgical treatments. There is need for non-hormonal, non-surgical treatment options for endometriosis, especially in those actively trying to conceive. Surgical management is frequently utilised; however, due to vast heterogeneity of the literature, the benefits of surgery are debated and appear to be dependent on surgeon, operating technique, symptoms, disease stage and multiple other factors [3, 9]. Recurrence is common after surgery, with increasing risk of recurrence over time. Treatment after surgery to prevent recurrence at present is largely reliant on suppression of menstruation, with limited efficacy [3].

Extracellular vesicles (EVs) are non-replicating particles delimited by a lipid bilayer that are released by all cells. EVs carry a variety of cargo involved in extracellular communication, altering the recipient cell phenotype [10]. EV is an umbrella term for different secreted particles secreted that differ by size and biogenesis, generally between 40 and 4000 nm in size, including apoptotic bodies, microvesicles and exosomes [11]. EVs contain bioactive cargo such as selective packaging of proteins, mRNA and miRNA species, which are reflective of the cell of origin and can provide insight into the physiological state of the tissue. EVs are abundant in bodily fluids making them prime candidates for use as “liquid biopsy”, having been successfully isolated from blood, urine and saliva [12]. There is hope that EVs may replace tissue diagnosis as a reliable and non-invasive diagnostic tool for many pathological processes [13].

This diagnostic and prognostic potential has led to an escalation in EV research in the past years [13]. As such, the International Society for Extracellular Vesicles (ISEV) was formed in 2012, providing reporting guidelines to ensure scientific rigour, reproducibility and validity of EV research. The minimal information for studies of extracellular vesicles (MISEV2018) guidelines proposed by the ISEV recommend all EV studies report detailed sample collection, EV isolation methodologies and EV characterization [14]. Knowledge of the role of EVs in endometriosis has rapidly expanded in recent years. These recent advances explore both diagnostic and therapeutic potential, as well as giving an ever-increasing understanding of the underlying pathophysiology. With longstanding multiple theories of endometriosis pathophysiology including Sampson’s theory of retrograde menstruation [15], metaplastic theory and others [16], EVs have provided a plausible connecting link between these hypotheses [16, 17]. It has been established that EVs released from the endometrium of people with endometriosis differ from those without endometriosis [18,19,20]; and retrograde flow of such EVs has been confirmed by their identification in peritoneal fluid [17]. It has been hypothesised that these EVs potentiate migration and implantation of endometrial cells shed during retrograde menstruation, as well as manipulating the immune response to avoid their destruction, bearing many similarities to their role in cancer [17]. Furthermore, EVs released from newly implanted ectopic endometrial cells (i.e. endometriosis lesions) have been identified, and hypothesised to drive angiogenesis and neurogenesis, creating vascular, proliferative and potentially painful lesions [17]. These processes are most plausible and relevant for peritoneal deposits, but may also play a role in endometriomas, deep infiltrating endometriosis lesions and even rarely reported distant lesions (e.g. lung, brain) [16]. As peritoneal endometriosis is the most challenging to diagnose without surgery, EVs hold potential as a non-surgical diagnostic tool for these patients. For EVs to be of diagnostic value, they need to be both unique to the process of endometriosis and easily detectable in non-invasive samples (such as blood, urine, cervical secretions or potentially even stool samples [21]). If both criteria could be met, especially for patients with disease not detectable on ultrasound, the need for laparoscopy to diagnose or exclude endometriosis could be avoided, and it may be possible to gain information about disease severity pre-operatively for those needing surgery. In addition, such EVs could provide therapeutic targets for non-hormonal and non-surgical treatment; potentially modifying disease progression and recurrence.

Here we present a systematic review of studies exploring EVs and their role in endometriosis, specifically addressing diagnostic and therapeutic potential and the contribution to the current understanding of pathophysiology. We explore which bodily fluids these EVs have been reported in, how the EVs were detected and their proposed function.

Methods

A systematic review was performed following the 2020 PRISMA guideline [22]. Five databases (Pubmed, Embase, Medline, Web of Science and Google Scholar [limited to 50 results by relevance]) were searched for keywords “endometriosis” and either “extracellular vesicles” or “exosomes” on 24/3/22 (see Additional File 1 for search strategy). No further studies were identified from citations during the literature review. Details of the protocol for this systematic review were registered on PROSPERO and can be accessed at https://www.crd.york.ac.uk/PROSPERO/display_record.php?ID=CRD42022302637.

Inclusion and exclusion criteria

Articles were included if they were in English, peer reviewed, and assessed samples of extracellular vesicles in subjects with endometriosis (with or without controls). Human and animal studies were included. Reviews and conference abstracts were excluded.

Data extraction

Studies were assessed for the following information: human or animal studies; source of bodily fluid to obtain EVs; isolation technique to identify EVs; target within EVs being studied; implications for diagnosis, treatment and pathogenesis of endometriosis; and limitations. Data from reports were extracted according to these headings (as shown in Table 1), with all three authors contributing independently to the data extraction and collation. No automation tools were required, and no further data was requested from included study authors.

Table 1 Key characteristics of studies of extracellular vesicles in endometriosis

Results

Search results

Twenty eight studies were included in the review. The initial search identified 274 articles of which 139 were unique. When applying inclusion criteria, 105 of these studies were excluded. A further six studies were excluded as they either did not specifically state that EVs were assessed, or did not specifically state that subjects had endometriosis. The flow of study identification is shown in Fig. 1.

Fig. 1
figure 1

Identification of studies via databases and register

Terminology

For the purposes of this review we use the term “endometrium” to imply eutopic endometrium; “endometriosis lesions” to describe any lesions outside the endometrium (commonly described as “ectopic endometrium” in the cited studies) and “endometrioma” to describe any endometriotic cysts on the ovary.

Much of the current EV literature uses inconsistent nomenclature, naming a subtype of EVs such as exosomes without demonstrating the biogenic origins of the EVs in the fractions isolated. Thus, this review will use the term ‘EV’ throughout.

Source of extracellular vesicles

Endometriosis related EVs were isolated from peritoneal fluid [18, 23, 27, 28, 30, 33, 46], endometrium [18,19,20, 25, 28, 29, 34, 35, 37, 38, 41, 42, 44, 47], endometriosis lesions [18, 19, 26, 28], endometriomas [25, 30, 34, 35, 39,40,41,42, 44, 47], cervicovaginal swabs (in a single monkey) [32], plasma [18, 31] and serum [18, 26, 28, 31, 34,35,36, 39, 43, 45, 47]. (Plasma is recommended by the ISEV over serum, due to the release of a significant amount of additional EVs from platelets during the clot formation when preparing serum [48]). Additionally, one study reported on EVs from umbilical cord derived stem cells, which were used in vitro as a potential therapeutic agent [24]. EVs were isolated from the conditioned media of primary ex vivo cultures in sixteen studies [19, 20, 24, 25, 28,29,30, 34, 35, 37, 38, 41, 42, 44, 46, 47].

Isolation and identification of extracellular vesicles

Isolation techniques varied between studies. Ultracentrifugation (UC) [19, 23, 25,26,27, 29, 30, 32, 37, 38, 41, 45, 46] and commercial kits [18, 20, 24, 28, 34,35,36, 39, 40, 42,43,44, 47] were the most commonly used methods (each used in thirteen studies) while two used size exclusion chromatography (SEC) [28, 33] and one was not reported [31]. Fifteen of twenty-eight studies (53%) fully characterised EVs according to the MISEV2018 guidlines, with the majority utilising western blot, electron microscopy and nanoparticle tracking analysis. Ten studies (36%) completed partial characterisation (did not fulfill all guidelines) and three (11%) did not report any EV characterisation.

Cargo and function of extracellular vesicles

The cargo of EVs were studied for their potential diagnostic utility, therapeutic utility or as a key element of the pathogenesis for endometriosis. Cargo studied include miRNAs involved with immunomodulation and cell proliferation [18, 20, 23, 43, 45, 46], angiogenesis [18, 19, 44] and fibrosis [41, 47]; proteins involved with angiogenesis [24, 28] and cell proliferation [25, 33]; lncRNAs involved with immunomodulation [30, 36], cell proliferation [36], cell migration [30, 35, 36] and angiogenesis [34]; and other targets including pseudogene LGMNP1 [39] and ectonucleotidases [40]. The specific cargo and their site of origin are listed in Fig. 2.

Fig. 2
figure 2

Extracellular vesicle cargo identified in subjects with endometriosis from different sites, with proposed function

Pathophysiology

Proposed roles of EV cargo in the pathogenesis of endometriosis are outlined in Fig. 2. Biopsies from the endometrium showed differences in EV cargo between people with and without endometriosis in five studies [18,19,20, 30, 42]. These included cargo involved in immunomodulation [30] and angiogenesis [19], and multiple miRNAs with as yet unclear function [18, 20]. Network analysis showed different expressions of miRNA, lncRNAs and mRNAs between groups [42].

EVs isolated from peritoneal fluid also showed differences between people with and without endometriosis in three studies [23, 33, 46]. Specifically, cargo were identified that play a role in cell migration [33], immunomodulation [23, 33, 46], cell proliferation [23] and angiogenesis [23, 33].

EVs isolated from endometriotic lesion biopsies contained cargo involved with cell migration [35], immunomodulation [26, 30, 39, 40] and angiogenesis [25, 28, 34, 44]. Several miRNAs with unclear function were also found to be present in levels significantly different from endometrial and peritoneal fluid samples in controls [18].

EVs isolated from plasma or serum showed differences between people with and without endometriosis in eight studies [18, 28, 34,35,36, 43, 45, 47]. Specifically, cargo were identified that play a role in cell migration [35], immunomodulation [18, 39, 43, 45], angiogenesis [18, 28, 34], fibrosis [47] and several lncRNAs with unclear function [36].

In addition, one study showed that EVs derived from mesenchymal stem cells may enhance migration of endometrial cells and therefore play a part in endometriosis [24]. Finally, one study showed differences in EVs derived from microbes in peritoneal fluid between women with endometriosis and controls, suggesting a role for microbiome in endometriosis aetiology [27]. The microbiome has been previously postulated to have an important role of the pathophysiology of endometriosis [21, 49]; EVs may provide a powerful tool to explore this further.

Diagnostic markers

Three studies reported on sensitivity and specificity of specific EV cargo as biomarkers [28, 36, 45]. One study of 48 patients and 21 controls reported serum EV derived VEGF-C as a biomarker with sensitivity 81.3% and specificity 71.4% for endometriosis [28]. Another study of 85 patients and 85 controls reported serum EV derived lncRNA RP3-399L15.2 as a biomarker with sensitivity 67% and specificity 98%; or using a combination of lncRNAs RP3-399L15.2 and CH507-513H4.6 a sensitivity of 80% and specificity of 85% [36]. A third study of 20 subjects and 20 controls reported a combination of serum EV derived miRNAs 320a and 22-3p as having sensitivity of 80% and specificity of 80% [45].

Additionally, a study of 52 patients and 21 controls reported serum derived pseudogene LGMNP1 having sensitivity 93% and specificity 76% in predicting recurrence within the four year study period, with a significant difference between subjects with primary and recurrent disease [39].

Extracellular vesicle derived therapeutic targets

Two studies utilised EVs derived from peritoneal macrophages as a potential therapeutic agent; demonstrating an ability to inhibit angiogenesis, migration and invasion of endometriosis in mouse models of endometriosis [29, 30]. One study utilised EV derived miR-214 to downregulate fibrosis in a mouse model of endometriosis [41]. Another study showed that EV derived miR-301a-3p was overexpressed in endometriosis lesions compared with serum from healthy controls; and that downregulation of this miRNA in a mouse model influenced macrophage activity [26].

Limitations

Specific limitations for each study are listed in Table 1.

Out of the twenty-six studies using patient tissue, thirteen reported endometriosis stage using the revised American Society of Reproductive Medicine (rASRM) classification system [18, 20, 23, 27, 28, 33, 36, 38, 41,42,43,44,45], four used the American Fertility Society (AFS) classification system [31, 34, 35, 39], and the remaining nine did not report the disease severity in the patient population [19, 24,25,26, 29, 30, 40, 46, 47]. Nine studies only included participants with endometriomas [20, 25, 34,35,36, 41, 42, 44, 47].

Other common limitations included small sample sizes, use of fetal bovine serum (FBS) for culture media supplementation without prior EV depletion [34, 39, 46] and unclear number of participants or samples used for assays.

Discussion

Whilst in its infancy, the use of EVs as diagnostic biomarkers or therapeutic targets in endometriosis has significant potential to improve the outcomes of people affected by this disease; additionally, several insights into the pathogenesis of the disease have been obtained. The recent excitement around EVs in cancer research has resulted in the development of strict reporting guidelines for researchers, with the next iteration of the MISEV guidelines to be released this year. As such the groundwork for investigating EVs in endometriosis is well established.

Using a systematic search protocol, we have identified and reviewed 28 studies that investigate EV cargo in endometriosis. Studies varied in their patient population, type of endometriosis, type of sample and EV isolation which created bias and difficulty in comparing studies; however, several insights into pathogenesis have been gained; diagnostic utility has been demonstrated; and therapeutic targets have been identified.

Limitations

There are multiple limitations in the included studies, which we broadly divide into clinical population limitations and laboratory processing limitations.

The clinical populations included in these studies were heterogeneous; with few studies reporting subject age and symptoms, and 65% reporting laparoscopic diagnosis and disease severity using accepted scoring systems. The limitation of current endometriosis scoring systems, especially with regards to clinical application, is well established; however in a research setting accepted scoring systems give some insight into the pathological extent of disease [50]. Nine studies only included subjects with endometriomas, and it is unclear at present whether EVs in this setting would differ from other manifestations of endometriosis.

With regards to laboratory processing bias, adherence to established MISEV standards was inconsistently achieved, limiting the reproducibility and reliability of many of the included studies. Complete EV characterisation is highly recommended to provide evidence of EV involvement when proposing a role in pathogenesis or EV cargo as biomarkers. The challenge of EV research is the heterogeneity of EV isolation methodologies creates difficulty in making meaningful comparisons across studies. Isolation methodology impacts on the composition of the EV fraction, with different size and density EVs and co-isolated contaminants favoured [51]. The lack of overlapping targets identified in any of the studies reviewed is likely a reflection of the diverse combinations of EV source, isolation techniques and methodologies used. Study reproducibility and the ability to validate study results is hindered by the absence of standardised sample processing and EV isolation methods.

The majority (57%) of the listed studies isolated EVs from ex vivo cell culture of primary stromal, mesenchymal or endometriosis samples. Cell culture purity or validation of cell type in these studies were not performed. The digestion of tissue and 2D culturing of primary cells in a variety of media (EV depleted or not) has a significant impact on the EV and EV cargo that is released [52]. Therefore, it is difficult to compare EV isolation from primary cell culture to those found directly in patient samples.

Diagnostic potential

There is a critical need for non-invasive diagnostic biomarkers in endometriosis. The lack of specific symptoms, limited understanding of the pathology of the disease and delay from symptom onset to diagnosis via invasive laparoscopy creates a serious clinical challenge in treating endometriosis. As cell signalling mediators which carry tissue/disease specific cargo, EVs hold the potential to be exploited as diagnostic biomarkers. EVs in peritoneal fluid and serum probably represent a combination of both diagnostic and prognostic categories; however, sampling peritoneal fluid is not particularly useful as a diagnostic tool as laparoscopy or other invasive procedures are required. Identifying the EV cargo derived from a combination of endometrium and endometriosis lesions in serum likely holds the greatest pragmatic potential as a non-invasive diagnostic biomarker and may provide insight into both “potential for disease” and “progression of disease”.

Four studies demonstrated that in samples taken from the endometrium, differences could be identified between people with and without endometriosis [18,19,20, 30]. As shown in Fig. 3, these differences may imply a susceptibility to developing endometriosis as molecules within these EVs appear to facilitate the development of endometriosis lesions by influencing cell migration and proliferation and immunomodulation. These EVs may contribute to a phenotype susceptible to developing endometriosis, and are unlikely to be affected by surgical resection of disease, however this is speculative and yet to be investigated.

Fig. 3
figure 3

Proposed role of extracellular vesicles (EVs) in the development of endometriosis lesions. EVs derived from the endometrium act on endometrial cells shed during retrograde menstruation, allowing migration, implantation and immunomodulation. EVs from newly implanted ectopic endometrial cells enhance pathogenesis with ongoing immunomodulation and cell proliferation, as well as angiogenesis and fibrosis, leading to the formation of endometriosis lesions

In contrast to this, EVs obtained from the peritoneal fluid and serum are likely to contain EVs derived from the endometrium in addition to EVs derived directly from endometriosis lesions, as well as numerous other sources. The diagnostic potential is therefore increased; however, reliable identification of the relevant EVs poses a more pronounced challenge. It could be assumed that analysis of EVs in these samples, in contrast to endometrial samples, would be markedly different after complete surgical excision of endometriosis; and therefore may bear promise as markers of current disease state. This could be exploited to detect recurrence, as was demonstrated in one small study [39].

Only 8 of the 28 studies systematically reviewed identified or discussed potential diagnostic markers, and only 4 of these measured AUC and/or sensitivity and specificity. These were serum RP3-399L15.2 [36] (Sn 67% and Sp 98%), serum VEGF [28] (Sn 81% Sp 71%), serum miR-22-3p and miR-320a [45] (AUC 0.883) and LGMNP1 [39] (as a marker for recurrence Sn 93% Sp 75%). Whilst promising, these studies have limited sample sizes (ranging from n = 20 to n = 86) and therefore further investigation and validation is required before clinical utility can be addressed.

Validation of these candidate diagnostic markers using clinically relevant means of isolation and quantification are also required. miR-22-3p and miR-320a were measured as biomarkers in EVs isolated using UC, while SEC was utilised when investigating EV-VEGF. While UC is the still the most widely used EV isolation methodology [53] and SEC generally yields higher purity EV fractions (depending on biofluid) [54], both have limited clinically utility due to specialist equipment requirements, typically time-consuming protocols and lack of scalability. Commercial kits tend to have short, straightforward protocols that are appealing for clinical use, however are plagued with high levels of non-EV contamination [55]. Therefore, the ideal means of EV isolation for clinical use may not yet have been developed; candidate biomarkers identified thus far may require future validation with a clinically implementable test.

Therapeutic potential

Many of the studies in this review used ex vivo cell culture techniques to investigate EV function in mediating macrophage polarisation, angiogenesis or proliferation. Therefore, the EV cargo identified in these studies were deemed potential therapeutic targets as they had some impact on endometriosis growth and inflammation.

Four studies using ex vivo cell culture techniques and mouse models were able to demonstrate disruption to the pathogenesis of endometriosis by targeting EVs therapeutically [26, 29, 30, 41]. As discussed above, the effect of ex vivo cell culture on EVs limits the reliability of such research; nonetheless there is promise and a myriad of potential targets for ongoing research to develop therapeutic agents. Significant advances need to be made before use in the clinical setting becomes conceivable.

If there is indeed a specific EV phenotype that facilitates development of endometriosis, it is plausible that therapeutic agents aimed at cargo of these specific EVs may be able to prevent disease or recurrence; however, the utility in treating already established lesions may be limited.

Recommendations for future research

As the diagnostic and therapeutic potential for endometriosis from EV research increases; we expect to see a larger focus on clinical aspects in future studies. Endometriosis is a vastly heterogeneous disease, with clinical symptoms ranging from asymptomatic through to severely debilitating; and surgical disease ranging from minimal to major pathological anatomical changes, with limited correlation between the two [56]. Detailed clinical data on study participants, their symptomatology and surgical data would add further understanding into how EVs differ across this incongruous disease spectrum.

While several studies have shown promising diagnostic potential for the use of EVs as a non-invasive diagnostic biomarker; validation of these studies in large populations is warranted; additionally, more detailed comparison between subjects with differing clinical and surgical severity would add further insight into the clinical utility of such tests. Future studies into diagnostic biomarkers should focus on using clinically accessible EV isolation methods, to maintain the relevance and translatability of the study.

The effect of surgical excision on endometriosis associated EVs is yet to be established. As outlined above, we hypothesise that some endometrium derived EVs may predispose to endometriosis, and that these would not be affected by surgical intervention; whilst EVs derived from endometriosis lesions would diminish significantly after surgery and therefore potentially act as a marker of recurrence. Only one study looked at recurrence [39] and would support this hypothesis, however, studies to specifically address this issue would provide further insight.

As shown in Fig. 2 above, there was very little crossover between identified EV targets across the 28 studies included in this review; and currently there are hundreds if not thousands of specific identified EV cargo from multiple bodily fluids that play a role in endometriosis. With such a wide number of comparisons across studies and relatively small participant numbers, it is likely that some false positives exist. There may be merit in future studies further validating currently identified targets rather than searching for new targets, such that meta-analyses may be performed in the future to bolster subject numbers. Identifying similar targets across multiple different bodily fluids may also provide further insight into their pathological relevance and clinical utility.

Finally, ongoing research into potential therapeutic EV targets is currently limited to animal models; however, as identification of relevant EV cargo and their pathological roles continues, further research into the disruption of endometriosis pathogenesis by targeting these EVs may provide the pathway for non-surgical non-hormonal treatment options.

Conclusion

Endometriosis is a chronic, inflammatory gynaecological disease that can have severe impacts on quality of life, placing burden on patients and the healthcare system. Due to the heterogeneous nature of endometriosis, diagnosis and treatment remain a significant clinical challenge. EVs are a relatively new field of biomedical research and have potential to be used as diagnostic tools in a range of diseases. EVs contribute to cell migration, implantation and immunomodulation in endometriosis; this can likely be exploited for diagnostic and prognostic information and may provide new therapeutic targets. We encourage further EV research in endometriosis to include rigorous reporting of both the patient population and technical methodology, with the goal of achieving clinical utility for diagnosis, prognosis and eventually treatment.

Availability of data and materials

Not applicable.

Abbreviations

AFS:

American Fertility Society

AUC:

Area under the curve

EV:

Extracellular vesicle

FBS:

Fetal bovine serum

ISEV:

International Society of Extracellular Vesicles

lncRNA:

Long non-coding RNA

miRNA:

Micro RNA

MISEV:

Minimal information for studies of extracellular vesicles

mRNA:

Messenger RNA

rASRM:

Revised American Society of Reproductive Medicine

SEC:

Size exclusion chromatography

UC:

Ultracentrifugation

References

  1. Ghiasi M, Kulkarni MT, Missmer SA. Is endometriosis more common and more severe than it was 30 years ago? J Minim Invasive Gynecol. 2020;27(2):452–61.

    Article  Google Scholar 

  2. Zondervan KT, Becker CM, Missmer SA. Endometriosis. N Engl J Med. 2020;382(13):1244–56.

    Article  CAS  Google Scholar 

  3. RANZCOG. Australian clinical practice guideline for the diagnosis and management of endometriosis. Melbourne. 2021.

  4. Ministry of Health. Diagnosis and management of endometriosis in New Zealand. Wellington: Ministry of Health; 2020.

    Google Scholar 

  5. Koninckx PR, Meuleman C, Demeyere S, Lesaffre E, Cornillie FJ. Suggestive evidence that pelvic endometriosis is a progressive disease, whereas deeply infiltrating endometriosis is associated with pelvic pain. Fertil Steril. 1991;55(4):759–65.

    Article  CAS  Google Scholar 

  6. Rowlands IJ, Abbott JA, Montgomery GW, Hockey R, Rogers P, Mishra GD. Prevalence and incidence of endometriosis in australian women: a data linkage cohort study. BJOG. 2021;128(4):657–65.

    Article  CAS  Google Scholar 

  7. Wykes CB, Clark TJ, Khan KS. Accuracy of laparoscopy in the diagnosis of endometriosis: a systematic quantitative review. BJOG. 2004;111(11):1204–12.

    Article  Google Scholar 

  8. Gubbels AL, Li R, Kreher D, Mehandru N, Castellanos M, Desai NA, et al. Prevalence of occult microscopic endometriosis in clinically negative peritoneum during laparoscopy for chronic pelvic pain. Int J Gynaecol Obstet. 2020;151(2):260–6.

    Article  Google Scholar 

  9. Bafort C, Beebeejaun Y, Tomassetti C, Bosteels J, Duffy JM. Laparoscopic surgery for endometriosis. Cochrane Database Syst Rev. 2020;10:CD011031.

    Google Scholar 

  10. Tetta C, Ghigo E, Silengo L, Deregibus MC, Camussi G. Extracellular vesicles as an emerging mechanism of cell-to-cell communication. Endocrine. 2013;44(1):11–9.

    Article  CAS  Google Scholar 

  11. Théry C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 2009;9(8):581–93.

    Article  Google Scholar 

  12. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–83.

    Article  CAS  Google Scholar 

  13. Esfandyari S, Elkafas H, Chugh RM, Park HS, Navarro A, Al-Hendy A. Exosomes as biomarkers for female Reproductive Diseases diagnosis and therapy. Int J Mol Sci. 2021;22(4):2165.

    Article  CAS  Google Scholar 

  14. Thery C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750.

    Article  Google Scholar 

  15. Sampson JA. The development of the implantation theory for the origin of peritoneal endometriosis. Am J Obstet Gynecol. 1940;40(4):549–57.

    Article  Google Scholar 

  16. Klemmt PAB, Starzinski-Powitz A. Molecular and Cellular Pathogenesis of Endometriosis. Curr Womens Health Rev. 2018;14(2):106–16.

    Article  CAS  Google Scholar 

  17. Shomali N, Hemmatzadeh M, Yousefzadeh Y, Soltani-Zangbar MS, Hamdi K, Mehdizadeh A, et al. Exosomes: emerging biomarkers and targets in folliculogenesis and endometriosis. J Reprod Immunol. 2020;142:103181.

    Article  CAS  Google Scholar 

  18. 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):e128846.

    Article  Google Scholar 

  19. Harp D, Driss A, Mehrabi S, Chowdhury I, Xu W, Liu D, et al. Exosomes derived from endometriotic stromal cells have enhanced angiogenic effects in vitro. Cell Tissue Res. 2016;365(1):187–96.

    Article  CAS  Google Scholar 

  20. Zhou W, Lian Y, Jiang J, Wang L, Ren L, Li Y, et al. Differential expression of microRNA in exosomes derived from endometrial stromal cells of women with endometriosis-associated infertility. Reprod Biomed Online. 2020;41(2):170–81.

    Article  Google Scholar 

  21. Laschke MW, Menger MD. The gut microbiota: a puppet master in the pathogenesis of endometriosis? Am J Obstet Gynecol. 2016;215(1):68 e1–4.

    Article  Google Scholar 

  22. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.

    Article  Google Scholar 

  23. 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. 2019;26(8):1130–8.

    Article  CAS  Google Scholar 

  24. Feng Y, Zhan F, Zhong Y, Tan B. Effects of human umbilical cord mesenchymal stem cells derived from exosomes on migration ability of endometrial glandular epithelial cells. Mol Med Rep. 2020;22(2):715–22.

    Article  CAS  Google Scholar 

  25. Hsu CY, Hsieh TH, Lin HY, Lu CY, Lo HW, Tsai CC, et al. Characterization and proteomic analysis of endometrial stromal cell-derived small Extracellular vesicles. J Clin Endocrinol Metab. 2021;106(5):1516–29.

    Article  Google Scholar 

  26. Huang Y, Zhu L, Li H, Ye J, Lin N, Chen M, et al. Endometriosis derived exosomal miR-301a-3p mediates macrophage polarization via regulating PTEN-PI3K axis. Biomed Pharmacother. 2022;147:112680.

    Article  CAS  Google Scholar 

  27. Lee SR, Lee JC, Kim SH, Oh YS, Chae HD, Seo H, et al. Altered composition of Microbiota in women with ovarian endometrioma: microbiome analyses of Extracellular vesicles in the peritoneal fluid. Int J Mol Sci. 2021;22(9):4608.

    Article  CAS  Google Scholar 

  28. Li WN, Hsiao KY, Wang CA, Chang N, Hsu PL, Sun CH, et al. Extracellular vesicle-associated VEGF-C promotes lymphangiogenesis and immune cells infiltration in endometriosis. Proc Natl Acad Sci U S A. 2020;117(41):25859–68.

    Article  CAS  Google Scholar 

  29. Li Q, Yuan M, Jiao X, Huang Y, Li J, Li D, et al. M1 macrophage-derived nanovesicles repolarize M2 macrophages for inhibiting the development of endometriosis. Front Immunol. 2021;12:707784.

    Article  CAS  Google Scholar 

  30. Liu T, Liu M, Zheng C, Zhang D, Li M, Zhang L. Exosomal lncRNA CHL1-AS1 derived from peritoneal Macrophages promotes the progression of endometriosis via the miR-610/MDM2 Axis. Int J Nanomedicine. 2021;16:5451–64.

    Article  Google Scholar 

  31. Munros J, Martinez-Zamora MA, Tassies D, Coloma JL, Torrente MA, Reverter JC, et al. Total circulating microparticle levels are increased in patients with deep infiltrating endometriosis. Hum Reprod. 2017;32(2):325–31.

    Article  CAS  Google Scholar 

  32. Muth DC, McAlexander MA, Ostrenga LJ, Pate NM, Izzi JM, Adams RJ, et al. Potential role of cervicovaginal extracellular particles in diagnosis of endometriosis. BMC Vet Res. 2015;11:187.

    Article  Google Scholar 

  33. Nazri HM, Imran M, Fischer R, Heilig R, Manek S, Dragovic RA, et al. Characterization of exosomes in peritoneal fluid of endometriosis patients. Fertil Steril. 2020;113(2):364–73. e2.

    Article  CAS  Google Scholar 

  34. Qiu JJ, Lin XJ, Zheng TT, Tang XY, Zhang Y, Hua KQ. The Exosomal Long Noncoding RNA aHIF is upregulated in serum from patients with endometriosis and promotes angiogenesis in endometriosis. Reprod Sci. 2019;26(12):1590–602.

    Article  CAS  Google Scholar 

  35. Qiu JJ, Lin YY, Tang XY, Ding Y, Yi XF, Hua KQ. Extracellular vesicle-mediated transfer of the lncRNA-TC0101441 promotes endometriosis migration/invasion. Exp Cell Res. 2020;388(1):111815.

    Article  CAS  Google Scholar 

  36. Shan S, Yang Y, Jiang J, Yang B, Yang Y, Sun F, et al. Extracellular vesicle-derived long non-coding RNA as circulating biomarkers for endometriosis. Reprod Biomed Online. 2022;44(5):923–33.

    Article  CAS  Google Scholar 

  37. Sun H, Li D, Yuan M, Li Q, Zhen Q, Li N, et al. Macrophages alternatively activated by endometriosis-exosomes contribute to the development of lesions in mice. Mol Hum Reprod. 2019;25(1):5–16.

    Article  CAS  Google Scholar 

  38. Sun H, Li D, Yuan M, Li Q, Li N, Wang G. Eutopic stromal cells of endometriosis promote neuroangiogenesis via exosome pathwaydagger. Biol Reprod. 2019;100(3):649–59.

    Article  Google Scholar 

  39. Sun SG, Guo JJ, Qu XY, Tang XY, Lin YY, Hua KQ, et al. The extracellular vesicular pseudogene LGMNP1 induces M2-like macrophage polarization by upregulating LGMN and serves as a novel promising predictive biomarker for ovarian endometriosis recurrence. Hum Reprod. 2022;37(3):447–65.

    Article  CAS  Google Scholar 

  40. Texido L, Romero C, Vidal A, Garcia-Valero J, Fernandez Montoli ME, Baixeras N, et al. Ecto-nucleotidases activities in the contents of ovarian endometriomas: potential biomarkers of endometriosis. Mediators Inflamm. 2014;2014:120673.

    Article  Google Scholar 

  41. Wu D, Lu P, Mi X, Miao J. Exosomal miR-214 from endometrial stromal cells inhibits endometriosis fibrosis. Mol Hum Reprod. 2018;24(7):357–65.

    CAS  Google Scholar 

  42. Wu J, Huang H, Huang W, Wang L, Xia X, Fang X. Analysis of exosomal lncRNA, miRNA and mRNA expression profiles and ceRNA network construction in endometriosis. Epigenomics. 2020;12(14):1193–213.

    Article  CAS  Google Scholar 

  43. Wu Y, Yuan W, Ding H, Wu X. Serum exosomal miRNA from endometriosis patients correlates with disease severity. Arch Gynecol Obstet. 2022;305(1):117–27.

    Article  CAS  Google Scholar 

  44. Wu J, Fang X, Huang H, Huang W, Wang L, Xia X. Construction and topological analysis of an endometriosis-related exosomal circRNA-miRNA-mRNA regulatory network. Aging. 2021;13(9):12607–30.

    Article  CAS  Google Scholar 

  45. Zhang L, Li H, Yuan M, Li D, Sun C, Wang G. Serum exosomal MicroRNAs as potential circulating biomarkers for endometriosis. Dis Markers. 2020;2020:2456340.

    Article  Google Scholar 

  46. Zhang L, Li HH, Yuan M, Li D, Wang GY. Exosomal mir-22-3p derived from peritoneal macrophages enhances proliferation, migration, and invasion of ectopic endometrial stromal cells through regulation of the SIRT1/NF-kappaB signaling pathway. Eur Rev Med Pharmacol Sci. 2020;24(2):571–80.

    CAS  Google Scholar 

  47. Zhang Y, Chang X, Wu D, Deng M, Miao J, Jin Z. Down-regulation of Exosomal miR-214-3p targeting CCN2 contributes to Endometriosis Fibrosis and the role of Exosomes in the horizontal transfer of miR-214-3p. Reprod Sci. 2021;28(3):715–27.

    Article  CAS  Google Scholar 

  48. Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, et al. Standardization of sample collection, isolation andanalysis methods in extracellular vesicle research. J Extracell Vesicles. 2013;27(2).

  49. Leonardi M, Hicks C, El-Assaad F, El-Omar E, Condous G. Endometriosis and the microbiome: a systematic review. BJOG. 2020;127(2):239–49.

    Article  CAS  Google Scholar 

  50. Johnson NP, Hummelshoj L, Adamson GD, Keckstein J, Taylor HS, Abrao MS, et al. World Endometriosis Society consensus on the classification of endometriosis. Hum Reprod. 2017;32(2):315–24.

    Article  Google Scholar 

  51. Brennan K, Martin K, FitzGerald SP, O’Sullivan J, Wu Y, Blanco A, et al. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci Rep. 2020;10(1):1039.

    Article  CAS  Google Scholar 

  52. Thippabhotla S, Zhong C, He M. 3D cell culture stimulates the secretion of in vivo like extracellular vesicles. Sci Rep. 2019;9(1):13012.

    Article  Google Scholar 

  53. Royo F, Thery C, Falcon-Perez JM, Nieuwland R, Witwer KW. Methods for separation and characterization of Extracellular vesicles: results of a Worldwide Survey performed by the ISEV Rigor and Standardization Subcommittee. Cells. 2020;9(9):1955.

    Article  CAS  Google Scholar 

  54. Yang Y, Wang Y, Wei S, Zhou C, Yu J, Wang G, et al. Extracellular vesicles isolated by size-exclusion chromatography present suitability for RNomics analysis in plasma. J Transl Med. 2021;19(1):104.

    Article  CAS  Google Scholar 

  55. Tian Y, Gong M, Hu Y, Liu H, Zhang W, Zhang M, et al. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J Extracell Vesicles. 2020;9(1):1697028.

    Article  CAS  Google Scholar 

  56. Vercellini P, Fedele L, Aimi G, Pietropaolo G, Consonni D, Crosignani PG. Association between endometriosis stage, lesion type, patient characteristics and severity of pelvic pain symptoms: a multivariate analysis of over 1000 patients. Hum Reprod. 2006;22(1):266–71.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

SS conceptualised the review and contributed to writing and editing of the manuscript. EP contributed to writing and editing of the draft manuscript. CH conceptualised the review, contributed to writing and editing of the manuscript, and provided supervision. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Claire E. Henry.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Scheck, S., Paterson, E.S.J. & Henry, C.E. A promising future for endometriosis diagnosis and therapy: extracellular vesicles - a systematic review. Reprod Biol Endocrinol 20, 174 (2022). https://doi.org/10.1186/s12958-022-01040-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12958-022-01040-y

Keywords