You are on page 1of 7

Biology of Reproduction, 2020, 102(6), 1153–1159

doi:10.1093/biolre/ioaa011
Review
Advance Access Publication Date: 22 January 2020

Review

The role of endometrial stem cells in the


pathogenesis of endometriosis and their

Downloaded from https://academic.oup.com/biolreprod/article/102/6/1153/5712138 by Universidad EIA user on 19 May 2021


application to its early diagnosis†
Yanli Liu1,2 , Zhiqin Zhang3 , Fen Yang1,2,4 , Hongmei Wang3 ,
Shengying Liang1,2 , Huiling Wang3 , Jun Yang3,* and Juntang Lin1,2,*
1 Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical

University, Xinxiang, China, 2 Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University,
Xinxiang, China, 3 The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China, and 4 College of
Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
∗ Correspondence: Jun Yang, The First Affiliated Hospital of Xinxiang Medical University, Jiankang Road, Weihui, Xinxiang,

Henan 453100, China. Tel: +86 13937335562; E-mail: 13937335562@163.com; Juntang Lin, Xinxiang Medical University, East
of JinSui Road, Xinxiang, Henan 453003, China. Tel: +86 0373 3831679; E-mail: linjtlin@126.com
† Grant Support: This study was supported by the National Natural Science Foundation of China (81671619 and U1804186),

the Xinxiang Foundation (20172DCG-03 and ZD17008) and the Xinxiang Medical University Foundation
(20172DCG-03 and 2017CXY-2-12).

Received 7 October 2019; Revised 21 December 2019; Accepted 16 January 2020

Abstract
Pelvic pain, infertility, and a high postoperative recurrence rate are associated with endometriosis
and adversely affect the physical and mental health of patients. Moreover, these factors place a
heavy burden on families and society. The identification of endometrial stem cells (EnSCs) in the
eutopic endometrium, menstrual blood, and ectopic lesions of women with endometriosis not only
provides new research objects in the context of endometriosis but also promotes and improves
our understanding of its pathogenesis. Furthermore, based on previous studies, we reasonably
suppose that dysfunctions of eutopic EnSCs play a critical role in the onset of endometriosis
and directly cause abnormalities in the endometrium; subsequently, retrograde menstruation
facilitates the delivery of abnormal endometrial tissues to the ovaries and pelvic cavity, where
they ectopically implant, grow, and form ectopic lesions. Additionally, as a chronically progressive
disease, there is a delay (3–11 years) from the first onset of symptoms to the diagnosis of
endometriosis. Therefore, the development of a method for early diagnosis with high sensitivity
and specificity is essential for endometriosis patients and has the potential to enable early treat-
ment, prevent endometriosis progression, and relieve pain in patients. Thus, focusing on EnSCs
will contribute to clarifying the potential pathogenesis of endometriosis and provide support for
the application of EnSCs as therapeutic and early diagnostic targets in endometriosis treatment.

Summary sentence
Focusing on endometrial stem cells (EnSCs) will contribute to clarifying the potential pathogenesis
of endometriosis and provide support for the application of EnSCs as therapeutic and early
diagnostic targets in endometriosis treatment.
Key words: endometriosis, endometrial stem cells, pathogenesis, early diagnosis

© The Author(s) 2020. Published by Oxford University Press on behalf of Society for the Study of Reproduction. All rights reserved.
For permissions, please e-mail: journals.permissions@oup.com 1153
1154 Y. Liu et al., 2020, Vol. 102, No. 6

Introduction Lang et al, also proposed an abnormal eutopic endometrium as the


pathogenesis of endometriosis, which suggests that a fundamental
Endometriosis refers to endometrial tissues (glands and stroma)
abnormality in the eutopic endometrium determines whether the
that are ectopically implanted in sites outside the endometrium and
endometrium can adhere, invade, and undergo angiogenesis outside
myometrium, and it is conservatively estimated that the incidence of
the uterine cavity; additionally, immune and endocrine factors, as
endometriosis in women of childbearing age is 10–15%. Currently,
auxiliary factors, promote the development of endometriosis [14,
the number of patients with endometriosis has exceeded 7 million
15]. The hypothesis of the abnormal eutopic endometrium partly
worldwide, and the incidence rate has been increasing annually [1, 2].
explains how retrograde menstruation can occur in most women of
Current studies have demonstrated that the clinical manifestations of
childbearing age but leads to endometriosis in only a small number
endometriosis include menstrual abnormalities, dysmenorrhoea, and
of women, which is an important supplement to and extension
pain during sexual intercourse, which are closely related to infertility.
of the theory of endometrial ectopic implantation caused by
Approximately, 50% of infertile women and 50–60% of women of

Downloaded from https://academic.oup.com/biolreprod/article/102/6/1153/5712138 by Universidad EIA user on 19 May 2021


retrograde menstruation. However, the exact mechanism leading to
childbearing age and adolescent women with various forms of pelvic
the abnormal activity of the eutopic endometrium in endometriosis
pain are definitively diagnosed with endometriosis [3, 4]. Gener-
patients is not yet clear. Therefore, the discovery of endometrial
ally, endometriosis manifests as diverse morphologies and extensive
stem cells (EnSCs) in the endometrium, menstrual blood, and ectopic
lesions. Although the lesions are not fatal, the presentation of cell
lesions is an important development for the existing hypotheses on
proliferation, infiltration, and recurrence are malignant biological
the pathogenesis of endometriosis and provides new targets for the
behaviors; additionally, the side effects caused by drugs and surgical
early diagnosis and treatment of endometriosis [16–18].
treatment and the high recurrence rate of endometriosis seriously
affect the physical and mental health of patients and place a heavy
burden to families and society [2, 5–7].
Endometrial stem cells
Conventionally, the endometrium undergoes periodic regeneration
and shedding approximately 500 times in healthy women during
Stem cell theory of endometriosis pathogenesis
their reproductive years, and the endometrium grows by approx-
To date, the pathogenesis of endometriosis is unclear. Hypotheses imately 5–7 mm per menstrual cycle, which strongly suggests the
regarding its pathogenesis mainly include the following: endometrial existence and involvement of stem cells in the periodic regenera-
ectopic implantation caused by retrograde menstruation, coelomic tion of the endometrium. As early as 2000, research demonstrated
metaplasia, embryonic cell rest, induction followed by lymphatic and that the stromal and glandular cells isolated from the menstrual
vascular dissemination, endocrinological factors, immunological and blood of healthy individuals can attach to the mesothelium and
genetic factors, and abnormalities in the eutopic endometrium [5, synergistically form endometriotic lesions only 5 days after being
7–10]. However, the events that are referred to in these hypotheses transplanted into nude mice [19]. A subsequent report also indicated
do not occur in isolation during the onset and development of that endometriotic lesions were likely to originate from the shedding
endometriosis. Regarding the pathogenic mechanism of endometrio- of cells or tissues with stemness properties from the basal layer
sis, the etiology of endometriosis is as follows: changes in genetic during menstruation [20]. Finally, a report in 2004 was the first
traits are present; an abnormal eutopic endometrium is the disease to demonstrate the existence of EnSCs with promising clonogenic
determinant; immune system abnormalities are prerequisites; and ability in the endometrium (endometrial epithelial progenitors and
retrograde menstruation is the bridge between the aforementioned stromal cells) [21].
latent factors. Together, these factors eventually lead to the onset of Currently, it is well known that EnSCs inhabit the basal
endometriosis. Simultaneously, coelomic metaplasia and induction layer of the endometrium, and can be successfully isolated from
followed by lymphatic and vascular dissemination are modifications endometrial tissues (eutopic endometrium and ectopic lesions of
and supplements to the pathogenesis of endometriosis [1, 7–10]. patients with endometriosis) after treatment with collagenase and
from menstrual blood after density gradient centrifugation or
direct treatment to lyse red blood cells. In addition to exhibiting
Ectopic endometrial implantation caused by high proliferative capacity and multipotency, EnSCs positively
retrograde menstruation and the abnormal eutopic express typical surface markers of mesenchymal stem cells, such
endometrium theory as CD29, CD44, CD73, CD90, and CD105, and are negative for
Currently, the most widely accepted theory of endometriosis is HLA-DR and hematopoietic cell markers, including CD34 and
endometrial ectopic implantation caused by retrograde menstrua- CD45 [22–24]. Further morphological, immunophenotyping, and
tion, as proposed by Sampson in 1921. Sampson’s theory proposed multipotency examinations have all confirmed that EnSCs meet
that endometrial glandular epithelial and mesenchymal cells enter the classic definition of mesenchymal stem cells [25]. Meanwhile,
the pelvic cavity by retrograde menstruation and are ectopically several reports have also demonstrated that EnSCs positively express
implanted into the ovary and the adjacent pelvic peritoneum, where embryonic stem cell markers, such as OCT-4, SOX2, and Nanog [24,
they continue to grow and spread to form ectopic lesions. However, 25]. In addition, the existence of EnSCs in the endometrium is rather
not all women with retrograde menstruation exhibit endometriosis stable, and menstruation and even menopause have no significant
[11]. influence on the existence of EnSCs [26].
Accumulative studies analyzing various processes, namely, gene With our increasing understanding of the heterogeneity of EnSCs
expression, proteomics, miRNA expression, hormone sensitivity, (including mesenchymal stem cells, endothelial progenitors, and
cell differentiation, and related cell signaling pathways, have epithelial progenitors) and different subtypes of EnSCs (constituting
revealed that there are significant differences between the eutopic CD146+ , SUSD2+ , ICAM1+ , and ABCG2+ subtypes) [26–29], the
endometrium of endometriosis patients and that of healthy subjects current results demonstrate that human EnSCs ectopically implanted
[12, 13]. Simultaneously, a group of Chinese scholars, namely, under the renal capsule of immunodeficient mice can form functional
Function of EnSCs in the pathogenesis of endometriosis, 2020, Vol. 102, No. 6 1155

endometrioid tissues and exhibit a response to exogenous estrogen the basal layer, suggesting dysfunction in eutopic EnSCs (EnSC-EM-
stimulation that is similar to that of the typical endometrium; Eu) in patients with endometriosis compared with healthy popula-
furthermore, the newly generated humanized blood vessels integrate tions (EnSC-Control). As expected, significant differences in mRNA
into the kidney tissues of mice to form a complete blood circulation expression between the EnSC-EM-Eu and EnSC-Control were subse-
system, and transplantation of a mixed population of EnSCs shows quently validated. The EnSC-EM-Eu and their differentiated fibrob-
enhanced endometrial tissue remodeling compared with the trans- lasts were all resistant to estrogen and without obvious decidualiza-
plantation of only a single subtype of EnSCs [28, 30]. Therefore, the tion [48]. Although Chan et al found no significant difference in the
abovementioned data strongly suggest the critical role of EnSCs in cloning efficiency between the EnSC-EM-Eu and EnSC-Control, the
the pathogenesis of endometriosis. authors still insisted that there was an undetectable difference in the
activity of these two types of EnSCs [49]. Simultaneously, compared
with healthy individuals, the expression of EnSC markers (Notch-

Downloaded from https://academic.oup.com/biolreprod/article/102/6/1153/5712138 by Universidad EIA user on 19 May 2021


EnSCs and endometriosis 1 and Numb) was significantly upregulated in the endometrium of
Current data have confirmed that angiogenesis and immune dys- patients with endometriosis based on a recent comparative analysis
regulation are important characteristics for the development of of a large number of cases, and the expression level of Notch-1
ectopic lesions in patients with endometriosis. Detection of pro- and Numb was positively correlated with endometriosis severity in
angiogenic factors and inflammatory factors in peritoneal fluid has clinical practice [50].
revealed that the levels of vascular endothelial growth factor (VEGF), Furthermore, several laboratories successfully isolated ectopic
epidermal growth factor, hepatocyte growth factor, and angiogenin EnSCs from the ectopic lesions of patients with endometriosis (EnSC-
in the peritoneal fluid of patients with endometriosis are significantly EM-Ec) and demonstrated that the in vitro migration, proliferation,
upregulated compared with the corresponding levels in the healthy and angiogenic capacities of EnSC-EM-Ec were all significantly
population, while inhibitors of angiogenesis, such as adiponectin improved compared with those of eutopic EnSCs. The enhanced
and IP-10, are downregulated; the degree of downregulation is invasion and angiogenic abilities of EnSC-EM-Ec in vivo were also
positively correlated with endometriosis severity [31, 32]. Simulta- demonstrated after their transplantation into immunodeficient mice
neously, a large number of studies have shown that the number and [51, 52]. Additionally, EnSC-EM-Ec exhibit distinct immune pheno-
activity of macrophages, neutrophils, T lymphocytes, and natural types from eutopic EnSCs by upregulating the expression of toll-like
killer cells in the abdominal cavity and ectopic lesions of patients receptors, collectin, pro-inflammatory factors, and migration, angio-
with endometriosis are significantly different from those of these genic and other related factors, thereby promoting the migration,
cell types in healthy populations. Abnormal lymphocytes not only proliferation and invasion abilities of eutopic EnSCs and increasing
directly promote the occurrence and development of ectopic lesions the degree of fibrosis in ectopic lesions [53, 54]. Furthermore,
by secreting a large number of bioactive factors (VEGF, IL-6 and IL- patients with endometriosis have more tissue shedding from the basal
8) but also induce a chronic inflammatory microenvironment in the layer during menstruation than do healthy individuals, and EnSC-
abdominal cavity and ectopic lesions, providing favorable conditions EM-Eu also show more chemotaxis to ectopic lesions, which can
for angiogenesis in ectopic lesions [33–35]. enrich EnSCs for the formation of ectopic lesions [20, 55].
Genetically, familial aggregation in endometriosis patients has Consequently, based on the published data, we reasonably
been confirmed [36], and the heritability of endometriosis is approx- speculated that the dysfunction of eutopic EnSCs (including the
imately 50%, which is estimated based on large studies of twins angiogenesis-promoting and immunomodulatory abilities) might
[37]. Because of this clear heritability, genome-wide association be a prerequisite for the occurrence of endometriosis, which
studies were performed, and nine previously reported and five novel directly leads to abnormalities in the eutopic endometrium; we also
loci significantly associated with endometriosis risk were identi- hypothesized that retrograde menstrual blood, as a bridge, enables
fied, which all harbor genes that function in sex steroid hormone the dysfunctional eutopic endometrium and EnSCs that become
pathways [38]; meanwhile, a dozen susceptibility regions were also detached during menstruation to ectopically implant in the ovary,
reported, but these regions only account for just over 4% of the pelvis, and other locations outside of uterus, and these implanted
heritability [7]. Furthermore, the role of environmental factors, cells can further proliferate and differentiate to form ectopic lesions
such as hypoxia [39], chronic inflammation [33], bacterial contam- under stimulation by a specific microenvironment (Figure 1).
ination (lipopolysaccharide) [40], endocrine disrupting chemicals
(metals/trace elements, dioxins, and other persistent organic pollu-
tants, nonpersistent chemicals) [41], and oxidative stress (reactive Application of EnSCs in endometriosis diagnosis
oxygen species) [42], have been demonstrated to contribute to and
promote the development of endometriosis. Conclusively, the impor- Current status of the clinical diagnosis
tant role of EnSCs in promoting angiogenesis and immunomod- of endometriosis
ulation has also been validated and supported by multiple thera- Because endometriosis is a progressive disease, patients usually expe-
peutic studies [25, 43–45], which allow us to reasonably speculate rience a delay from the onset of the first symptoms to the definitive
that EnSCs are likely to induce dysfunctional angiogenesis and diagnosis of endometriosis (3–11 years), namely, the endometriosis
immunomodulation via the above-mentioned changes in genetic diagnostic delay; for patients with dysmenorrhoea starting from
or environmental factors, which can promote ectopic implantation menarche, the time of diagnostic delay is even longer [5, 8]. Thus
during retrograde menstruation, resulting in the formation of ectopic far, routine endometriosis diagnostic methods, mainly including
lesions. laparoscopic findings of ectopic lesions and pathological biopsy
Previous reports [46, 47] have confirmed that the activity of examinations, as well as assisted laboratory examinations and pelvic
the eutopic endometrium in endometriosis patients is significantly ultrasound or abdominal computed tomography examinations, can
different from that in the healthy population and that the eutopic confirm the diagnosis of endometriosis and the severity of the
endometrium periodically regenerates from the EnSCs that reside in disease. Nevertheless, the diagnostic delay often results in delayed
1156 Y. Liu et al., 2020, Vol. 102, No. 6

Downloaded from https://academic.oup.com/biolreprod/article/102/6/1153/5712138 by Universidad EIA user on 19 May 2021


Figure 1. The hypothesis of this study. Dysfunctional eutopic EnSCs play a critical role in the onset of endometriosis by directly causing eutopic endometrium
abnormalities; subsequently, retrograde menstruation delivers dysfunctional eutopic endometrial tissue and EnSCs that are detached during menstruation to
ectopic sites such as the ovaries and pelvic cavity, where they implant and form ectopic lesions.

treatment, which can cause disease deterioration. Laparoscopy, indicator need to be improved [57, 58]. Therefore, CA125 has been
which is the gold standard for endometriosis diagnosis, still has combined with other indicators (including CA199, anti-endometrial
some shortcomings, including trauma, high cost, potential risks antibodies, aldehyde dehydrogenase 1, prolactin, epididymal pro-
after anesthesia, and visceral tissue and vascular injury; moreover, tein 4, related inflammatory factors, and endometrial aromatase)
laparoscopy may lead to the missed diagnosis of some deep or to address its insufficiency as a single indicator and to improve
invisible lesions, which limits its extensive application in the early the sensitivity and specificity of early endometriosis diagnosis. In
diagnosis and treatment of endometriosis [2, 56]. addition, based on data such as serum proteomics, peripheral blood
Generally, with their advantages of being noninvasive and objec- miRNAs, and urine markers, early endometriosis diagnosis has
tive, serological markers are more easily accepted by patients and been extensively investigated, and several promising indicators have
gynecologists than laparoscopy, but there is still no effective or been identified and tested [59–62]. Although the aforementioned
unique serological marker for the early diagnosis of endometriosis diagnostic methods have positive effects on the clinical diagnosis
in clinical practice. To date, serum CA125 has been the most widely and recurrence of endometriosis, there are still many shortcomings
used diagnostic indicator for endometriosis in clinical practice, and a regarding the early diagnosis and risk prediction of potential patients
large amount of clinical data have demonstrated that the sensitivity with endometriosis.
of the CA125 indicator is positively correlated with the severity
of endometriosis, especially for patients with stages III and IV
endometriosis; however, for some patients with mild endometriosis, Potential application of menstrual blood derived
the serum CA125 indicator levels are normal, indicating that serum EnSCs in early endometriosis diagnosis
CA125 alone is insufficient for the early diagnosis of endometriosis Previous studies [46, 47] have focused on comparative analyses of
and that the sensitivity and specificity of serum CA125 as a single the differential expression of related genes and proteins in the eutopic
Function of EnSCs in the pathogenesis of endometriosis, 2020, Vol. 102, No. 6 1157

endometrium of patients with endometriosis, in ectopic lesions and in diagnostic strategies based on MenSCs can be used to improve early
the eutopic endometrium of healthy people to elucidate the potential detection and risk evaluations of potential patients with endometrio-
pathogenesis of endometriosis at the tissue level. However, there are sis, and early interventions can be initiated to prevent or slow disease
many types of cells involved in the formation of the endometrium progression.
and ectopic lesions, and these cells are synergistically regulated by a
variety of endocrine and immune signals [33, 63]; moreover, there are
significant variations in the endometrium at different physiological Conflict of Interests
stages (proliferation, secretion, and menstruation) [64, 65], which
seriously affect the reliability of research results and even result in The authors declare that there is no conflict of interest regarding the
conflicting conclusions. Therefore, taking the EnSCs implanted in publication of this paper.
eutopic and ectopic lesions as the research object can effectively

Downloaded from https://academic.oup.com/biolreprod/article/102/6/1153/5712138 by Universidad EIA user on 19 May 2021


reduce the number of complex variables involved in exploring the
pathogenesis of endometriosis at the tissue level and ensure the
References
validity of the results. At the end of 2007, menstrual blood-derived
stromal/stem cells (MenSCs) were successfully isolated, and their 1. Brown J, Farquhar C. An overview of treatments for endometriosis. JAMA
biological characteristics were consistent with the standards of mes- 2015; 313:296–297.
2. Maggiore ULR, Ferrero S, Mangili G, Bergamini A, Inversetti A, Giorgione
enchymal stem cells established by the International Cell Therapy
V, Viganò P, Candiani M. A systematic review on endometriosis during
Association [23, 66]. Our previous findings have directly proven
pregnancy: diagnosis, misdiagnosis, complications and outcomes. Hum
that MenSCs reside in the deciduous endometrium, which suggests Reprod Update 2015; 22:70–103.
that MenSCs isolated by non-invasive methods have biological char- 3. Goldstein DP, deCholnoky C, Emans SJ, Leventhal JM. Laparoscopy in
acteristics similar to those of EnSCs [24]. Recently, MenSCs have the diagnosis and management of pelvic pain in adolescents. J Reprod
received extensive attention in view of their advantages, namely, their Med 1980; 24:251–256.
abundance and high proliferative capacity, as well as the ease and 4. Eskenazi B, Warner ML. Epidemiology of endometriosis. Obstet Gynecol
non-invasiveness of MenSC sample collection methods [25, 43–45]. Clin N Am 1997; 24:235–258.
As expected, the application of MenSCs has resulted in satisfactory 5. Bulun SE. Endometriosis. N Engl J Med 2009; 360:268–279.
therapeutic effects in many animal disease models and clinical studies 6. Maggiore ULR, Ferrero S, Candiani M, Somigliana E, Viganò P, Vercellini
P. Bladder endometriosis: a systematic review of pathogenesis, diagnosis,
[67, 68].
treatment, impact on fertility, and risk of malignant transformation. Eur
To date, the exploration of differences between EnSC-EM-Eu and
Urol 2017; 71:790–807.
EnSC-Control is still in the preliminary stage. However, published 7. Borghese B, Zondervan KT, Abrao MS, Chapron C, Vaiman D. Recent
data have confirmed that cell detachment in the basal layer of the insights on the genetics and epigenetics of endometriosis. Clin Genet 2017;
endometrium during menstruation is significantly higher in patients 91:254–264.
with endometriosis than in healthy individuals and that the detached 8. Giudice LC, Kao LC. Endometriosis, Lancet 2004; 364:1789–1799.
cells involved in retrograde menstruation are likely to provide a 9. Greene AD, Lang SA, Kendziorski JA, Sroga-Rios JM, Herzog TJ, Burns
sufficient cell source for the formation of potential ectopic lesions KA. Endometriosis: where are we and where are we going? Reproduction
[20]. Furthermore, significant morphological differences have been 2016; 152:R63–R78.
observed between MenSCs isolated from patients with endometrio- 10. Chapron C, Marcellin L, Borghese B. Rethinking mechanisms, diag-
nosis and management of endometriosis. Nat Rev Endocrinol 2019;
sis and those isolated from healthy individuals; compared with
15:666–682.
the MenSCs of healthy individuals, the MenSCs of endometriosis
11. Sampson JA. Metastatic or embolic endometriosis, due to the menstrual
patients had CD9, CD10, and CD29 expression levels that were dissemination of endometrial tissue into the venous circulation. Am J
significantly upregulated, proliferation and invasion capacities that Pathol 1927; 3:93–109.
were significantly enhanced, and an immunomodulation ability that 12. Aghajanova L, Tatsumi K, Horcajadas JA, Zamah AM, Esteban FJ,
was significantly downregulated [69]. Herndon CN, Conti M, Giudice LC. Unique transcriptome, pathways, and
Thus, based on the above results, we contend that MenSCs can be networks in the human endometrial fibroblast response to progesterone
used as a promising target and that further investigation of the dif- in endometriosis. Biol Reprod 2011; 84:801–815.
ferences in the biological characteristics of MenSCs between patients 13. Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard M, Nezhat CR,
with endometriosis and healthy individuals should be performed to Lessey BA, Giudice LC. Gene expression analysis of endometrium reveals
progesterone resistance and candidate susceptibility genes in women with
screen potential biomarkers (genes or proteins) that underlie the
endometriosis. Endocrinology 2007; 148:3814–3826.
differences between the MenSCs mentioned above. Simultaneously,
14. Li X, Gong X, Zhu L, Leng J, Fan Q, Sun D, Lang J, Fan Y.
technological development (high-throughput sequencing and protein Stretch magnitude- and frequency-dependent cyclooxygenase 2 and
assays) can promote and optimize the combination of these poten- prostaglandin E2 up-regulation in human endometrial stromal cells:
tial biomarkers, which are promising for use as early noninvasive possible implications in endometriosis. Exp Biol Med (Maywood) 2012;
endometriosis diagnostic strategies. Additionally, with expansion of 237:1350–1358.
the sample size, the diagnostic parameters can be gradually improved 15. Sha G, Zhang Y, Zhang C, Wan Y, Zhao Z, Li C, Lang J. Elevated levels of
and optimized. gremlin-1 in eutopic endometrium and peripheral serum in patients with
endometriosis. Fertil Steril 2009; 91:350–358.
16. Cousins FL, Xiao L, Gargett CE. Adult stem cells in the pathogenesis
Conclusion and treatment of endometriosis. J Endometr Pelvic Pain Disord 2017;
9:223–231.
In summary, the in-depth study of EnSCs can improve the stem cell 17. Djokovic D, Calhaz-Jorge C. Somatic stem cells and their dysfunction in
theory regarding the pathogenesis of endometriosis, partly explain- endometriosis. Front Surg 2015; 1:1–8.
ing the onset and development of ectopic lesions at the cellu- 18. Cousins FL, O DF, Gargett CE. Endometrial stem/progenitor cells and
lar level and providing supplements for the existing pathogene- their role in the pathogenesis of endometriosis. Best Pract Res Clin Obstet
sis of endometriosis. Furthermore, early noninvasive endometriosis Gynaecol 2018; 50:27–38.
1158 Y. Liu et al., 2020, Vol. 102, No. 6

19. Nisolle M, Casanas-Roux F, Donnez J. Early-stage endometriosis: adhe- 42. Donnez J, Binda MM, Donnez O. Oxidative stress in the pelvic cavity
sion and growth of human menstrual endometrium in nude mice. Fertil and its role in the pathogenesis of endometriosis. Fertil Steril 2016;
Steril 2000; 74:306–312. 106:1011–1017.
20. Leyendecker G, Herbertz M, Kunz G, Mall G. Endometriosis results from 43. Ulrich D, Muralitharan R. Toward the use of endometrial and menstrual
the dislocation of basal endometrium. Hum Reprod 2002; 17:2725–2736. blood mesenchymal stem cells for cell-based therapies. Expert Opin Biol
21. Chan RW, Schwab KE, Gargett CE. Clonogenicity of human endometrial Ther 2013; 13:1387–1400.
epithelial and stromal cells. Biol Reprod 2004; 70:1738–1750. 44. Lv H, Hu Y, Cui Z. Human menstrual blood: a renewable and sustainable
22. Gargett CE, Schwab KE. Endometrial stem/progenitor cells: the first 10 source of stem cells for regenerative medicine. Stem Cell Res Ther 2018;
years. Hum Reprod Update 2016; 22:137–163. 9:325.
23. Meng X, Ichim TE, Zhong J, Rogers A, Yin Z, Jackson J, Wang H, Ge W, 45. Liu Y, Niu R, Li W, Lin J, Stamm C, Steinhoff G, Ma N. Therapeutic
Bogin V, Chan KW, Thébaud B, Riordan NH. Endometrial regenerative potential of menstrual blood-derived endometrial stem cells in cardiac
cells: A novel stem cell population. J Transl Med 2007; 5:57–66. diseases. Cell Mol Life Sci 2019; 76:1681–1695.

Downloaded from https://academic.oup.com/biolreprod/article/102/6/1153/5712138 by Universidad EIA user on 19 May 2021


24. Liu Y, Niu R, Yang F, Yan Y, Liang S, Sun Y, Shen P, Lin J. Biological 46. Haikalis ME, Wessels JM, Leyland NA, Agarwal SK, Foster WG.
characteristics of human menstrual blood-derived endometrial stem cells. MicroRNA expression pattern differs depending on endometriosis lesion
J Cell Mol Med 2018; 22:1627–1639. type. Biol Reprod 2018; 98:623–633.
25. Chen L, Qu J, Xiang C. The multi-functional roles of menstrual blood- 47. Ferrero S. Proteomics in the diagnosis of endometriosis: opportunities and
derived stem cells in regenerative medicine. Stem Cell Res Ther 2019; 10:1. challenges. Proteomics Clin Appl 2019; 13:e1800183.
26. Ulrich D, Tan KS, Deane J, Schwab K, Cheong A, Rosamilia A, Gargett 48. Barragan F, Irwin JC, Balayan S, Erikson DW, Chen JC, Houshdaran S,
CE. Mesenchymal stem/stromal cells in post-menopausal endometrium. Piltonen TT, Spitzer TL, George A, Rabban JT, Nezhat C, Giudice LC.
Hum Reprod 2014; 29:1895–1905. Human endometrial fibroblasts derived from mesenchymal progenitors
27. Schwab KE, Gargett CE. Co-expression of two perivascular cell markers inherit progesterone resistance and acquire an inflammatory pheno-
isolates mesenchymal stem-like cells from human endometrium. Hum type in the endometrial niche in endometriosis. Biol Reprod 2016; 94,
Reprod 2007; 22:2903–2911. 118.
28. Masuda H, Anwar SS, Bühring HJ, Rao JR, Gargett CE. A novel marker 49. Chan RWS, Ng EHY, Yeung WSB. Identification of cells with colony-
of human endometrial mesenchymal stem-like cells. Cell Transplant 2012; forming activity, self-renewal capacity and multipotency in ovarian
21:2201–2214. endometriosis. Am J Pathol 2011; 178:2832–2844.
29. Tsuji S, Yoshimoto M, Takahashi K, Noda Y, Nakahata T, Heike T. Side 50. Schüring AN, Dahlhues B, Korte A, Kiesel L, Titze U, Heitkötter B,
population cells contribute to the genesis of human endometrium. Fertil Ruckert C, Götte M. The endometrial stem cell markers notch-1 and
Steril 2008; 90:1528–1537. numb are associated with endometriosis. Reprod Biomed Online 2018;
30. Masuda H, Maruyama T, Hiratsu E, Yamane J, Iwanami A, Nagashima 36:294–301.
T, Ono M, Miyoshi H, Okano HJ, Ito M, Tamaoki N, Nomura T et 51. Canosa S, Moggio A, Brossa A, Pittatore G, Marchino GL, Leoncini S,
al. Noninvasive and real-time assessment of reconstructed functional Benedetto C, Revelli A, Bussolati B. Angiogenic properties of endometrial
human endometrium in NOD/SCID/gamma c (null) immunodeficient mesenchymal stromal cells in endothelial co-culture: an in vitro model of
mice. PNAS 2007; 104:1925–1930. endometriosis. Mol Hum Reprod 2017; 23:187–198.
31. Laschke MW, Menger MD. Basic mechanisms of vascularization in 52. Kao AP, Wang KH, Chang CC, Lee JN, Long CY, Chen HS, Tsai
endometriosis and their clinical implications. Hum Reprod Update 2018; CF, Hsieh TH, Tsai EM. Comparative study of human eutopic and
24:207–224. ectopic endometrial mesenchymal stem cells and the development
32. Rakhila H, Al-Akoum M, Bergeron ME, Leboeuf M, Lemyre M, Akoum of an in vivo endometriotic invasion model. Fertil Steril 2011; 95:
A, Pouliot M. Promotion of angiogenesis and proliferation cytokines 1308–1315.
patterns in peritoneal fluid from women with endometriosis. J Reprod 53. Li J, Dai Y, Zhu H, Jiang Y, Zhang S. Endometriotic mesenchymal stem
Immunol 2016; 116:1–6. cells significantly promote fibrogenesis in ovarian endometrioma through
33. Izumi G, Koga K, Takamura M, Makabe T, Satake E, Takeuchi A, the Wnt/β-catenin pathway by paracrine production of TGF-β1 and
Taguchi A, Urata Y, Fujii T, Osuga Y. Involvement of immune cells in the Wnt1. Hum Reprod 2016; 31:1224–1235.
pathogenesis of endometriosis. J Obstet Gynaecol Re 2018; 44:191–198. 54. Nair ARKG, Pandit H, Warty N, Madan T. Endometriotic mesenchymal
34. Jiang L, Yan Y, Liu Z, Wang Y. Inflammation and endometriosis. Front stem cells exhibit a distinct immune phenotype. Int Immunol 2015;
Biosci 2016; 21:941–948. 27:195–204.
35. Ahn SH, Khalaj K, Young SL, Lessey BA, Koti M, Tayade C. Immune- 55. Cheng Y, Li L, Wang D, Guo Q, He Y, Liang T, Sun L, Wang X, Cheng Y,
inflammation gene signatures in endometriosis patients. Fertil Steril 2016; Zhang G. Characteristics of human endometrium-derived mesenchymal
106:1420–1431. stem cells and their tropism to endometriosis. Stem Cells Int 2017;
36. Stefansson H, Geirsson RT, Steinthorsdottir V, Jonsson H, Manolescu A, 2017:4794827.
Kong A, Ingadottir G, Gulcher J. Genetic factors contribute to the risk of 56. Parasar P, Ozcan P, Terry KL. Endometriosis: epidemiology, diagnosis and
developing endometriosis. Hum Reprod 2002; 17:555–559. clinical management. Curr Obstet Gynecol Rep 2017; 6: 34–41.
37. Treloar SA, O’Connor DT, O’Connor VM. Genetic influences on 57. Hirsch M, Duffy JMN, Deguara CS, Davis CJ, Khan KS. Eur J Obstet
endometriosis in an Australian twin sample. sueT@qimr.edu.au. Fertil Gynecol Reprod Biol 2016; 210:102–107.
Steril 1999; 71:701–710. 58. Baggio S, Zecchin A, Pomini P, Zanconato G, Genna M, Motton M, Mon-
38. Sapkota Y, Steinthorsdottir V, Morris AP, Fassbender A, Rahmioglu N, temezzi S, Franchi M. The role of computed tomography colonography in
Vivo D, Buring JE, Zhang F, Edwards TL, Jones SOD, Peterse D et detecting bowel involvement in women with deep infiltrating endometrio-
al. Meta-analysis identifies five novel loci associated with endometriosis sis: comparison with clinical history, serum CA125, and transvaginal
highlighting key genes involved in hormone metabolism. Nat Commun sonography. J Comput Assist Tomogr 2016; 40:886–891.
2017; 8:15539. 59. Ahn SH, Singh V, Tayade C. Biomarkers in endometriosis: challenges and
39. Wu MH, Hsiao KY. Hypoxia: the force of endometriosis. J Obstet opportunities. Fertil Steril 2017; 107:523–532.
Gynaecol Res 2019; 45:532–541. 60. May KE, Villar J, Kirtley S, Kennedy SH, Becker CM. Endometrial
40. Khan KN, Fujishita A, Hiraki K, Kitajima M, Nakashima M, Fushiki alterations in endometriosis: a systematic review of putative biomarkers.
S, Kitawaki J. Bacterial contamination hypothesis: a new concept in Hum Reprod Update 2011; 17:637–653.
endometriosis. Reprod Med Biol 2018; 17:125–133. 61. Gupta D, Hull ML, Fraser I, Miller L, Bossuyt PM, Johnson N, Nisenblat
41. Smarr MM, Kannan K. Endocrine disrupting chemicals and endometrio- V. Endometrial biomarkers for the non-invasive diagnosis of endometrio-
sis. Fertil Steril 2016; 106:959–966. sis. Cochrane Database Syst Rev 2016; 4:CD012165.
Function of EnSCs in the pathogenesis of endometriosis, 2020, Vol. 102, No. 6 1159

62. Nothnick WB. MicroRNAs and endometriosis: distinguishing drivers 67. Mutlu L, Hufnagel D, Taylor HS. The endometrium as a source of
from passengers in disease pathogenesis. Semin Reprod Med 2017; mesenchymal stem cells for regenerative medicine. Biol Reprod 2015;
35:173–180. 92:138.
63. Patel BG, Lenk EE, Lebovic DI, Shu Y, Yu J, Taylor RN. Pathogenesis 68. Domnina A, Novikova P, Obidina J, Fridlyanskaya I, Alekseenko
of endometriosis: interaction between endocrine and inflammatory path- L, Kozhukharova I, Lyublinskaya O, Zenin V, Nikolsky N. Human
ways. Best Pract Res Clin Obstet Gynaecol 2018; 50:50–60. mesenchymal stem cells in spheroids improve fertility in model ani-
64. Silverberg SG. The endometrium. Arch Pathol Lab Med 2007; mals with damaged endometrium. Stem Cell Res Ther 2018; 9:
131:372–382. 50.
65. Munro SK, Farquhar CM, Mitchell MD. Epigenetic regulation of 69. Nikoo S, Ebtekar M, Jeddi-Tehrani M, Shervin A, Bozorgmehr M,
endometrium during the menstrual cycle. Mol Hum Reprod 2010; Vafaei S, Kazemnejad S, Zarnani AH. Menstrual blood-derived stromal
16:297–310. stem cells from women with and without endometriosis reveal different
66. Patel AN, Park E, Kuzman M, Benetti F, Silva FJ, Allickson JG. Multipo- phenotypic and functional characteristics. Mol Hum Reprod 2014; 20:

Downloaded from https://academic.oup.com/biolreprod/article/102/6/1153/5712138 by Universidad EIA user on 19 May 2021


tent menstrual blood stromal stem cells: isolation, characterization, and 905–918.
differentiation. Cell Transplant 2008; 17:303–311.

You might also like