You are on page 1of 33

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/332475915

Endometriosis

Article  in  Endocrine Reviews · April 2019


DOI: 10.1210/er.2018-00242

CITATIONS READS

279 3,441

10 authors, including:

Christia Angela Sison Kaoru Miyazaki


Children's Hospital Los Angeles Hara Medical Clinic
5 PUBLICATIONS   333 CITATIONS    41 PUBLICATIONS   973 CITATIONS   

SEE PROFILE SEE PROFILE

Lia Bernardi Shimeng Liu


Northwestern University Northwestern University
58 PUBLICATIONS   1,260 CITATIONS    21 PUBLICATIONS   519 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Endometriosis View project

surgical education scholars View project

All content following this page was uploaded by Christia Angela Sison on 29 July 2019.

The user has requested enhancement of the downloaded file.


REVIEW

Endometriosis

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
Serdar E. Bulun,1 Bahar D. Yilmaz,1 Christia Sison,1 Kaoru Miyazaki,1 Lia Bernardi,1
Shimeng Liu,1 Amanda Kohlmeier,1 Ping Yin,1 Magdy Milad,1 and JianJun Wei1,2

1
Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago,
Illinois 60611; and 2Department of Pathology, Feinberg School of Medicine, Northwestern University,
Chicago, Illinois 60611
ORCiD numbers: 0000-0003-0850-8637 (S. E. Bulun).

ABSTRACT Pelvic endometriosis is a complex syndrome characterized by an estrogen-dependent chronic inflammatory process that
affects primarily pelvic tissues, including the ovaries. It is caused when shed endometrial tissue travels retrograde into the lower
abdominal cavity. Endometriosis is the most common cause of chronic pelvic pain in women and is associated with infertility. The
underlying pathologic mechanisms in the intracavitary endometrium and extrauterine endometriotic tissue involve defectively
programmed endometrial mesenchymal progenitor/stem cells. Although endometriotic stromal cells, which compose the bulk of
endometriotic lesions, do not carry somatic mutations, they demonstrate specific epigenetic abnormalities that alter expression of key
transcription factors. For example, GATA-binding factor-6 overexpression transforms an endometrial stromal cell to an endometriotic
phenotype, and steroidogenic factor-1 overexpression causes excessive production of estrogen, which drives inflammation via
pathologically high levels of estrogen receptor-b. Progesterone receptor deficiency causes progesterone resistance. Populations of
endometrial and endometriotic epithelial cells also harbor multiple cancer driver mutations, such as KRAS, which may be associated with
the establishment of pelvic endometriosis or ovarian cancer. It is not known how interactions between epigenomically defective stromal
cells and the mutated genes in epithelial cells contribute to the pathogenesis of endometriosis. Endometriosis-associated pelvic pain is
managed by suppression of ovulatory menses and estrogen production, cyclooxygenase inhibitors, and surgical removal of pelvic lesions,
and in vitro fertilization is frequently used to overcome infertility. Although novel targeted treatments are becoming available, as
endometriosis pathophysiology is better understood, preventive approaches such as long-term ovulation suppression may play a critical
role in the future. (Endocrine Reviews 40: 1048 – 1079, 2019)

Definition of Endometriosis the key biologic driver of inflammation, however,


makes endometriosis unique (–).

A dvances made during the last two decades have


revealed endometriosis as a complex clinical
syndrome characterized by an estrogen-dependent
The classical definition of endometriosis is the
surgical detection of endometrial tissue outside of the
uterine cavity (); however, this narrow anatomic
chronic inflammatory process that affects primarily definition has proven insufficient to explain the nat-
pelvic tissues, including the ovaries (, ). Endome- ural history of endometriosis, the full spectrum of its
ISSN Print: 0163-769X triosis is the most common cause of chronic pelvic clinical features, the frequent recurrence of its
ISSN Online: 1945-7189 pain in reproductive-age women and is strongly linked symptoms, the underlying molecular pathophysiology,
Printed: in USA to persistent episodes of ovulation, menstruation, and or its responsiveness to currently available manage-
Copyright © 2019
cycling steroid hormones (, ). Its multifactorial ment modalities (, , , ). Recently, the definition of
Endocrine Society
Received: 27 September 2018
etiology and high prevalence resemble other chronic endometriosis has evolved to one that is more patient-
Accepted: 8 April 2019 inflammatory disorders associated with pain, such as focused and takes into account the cellular and mo-
First Published Online: inflammatory bowel disease and gastroesophageal lecular origins of the disease; its natural history from
17 April 2019 reflux disorder (, ). Its dependence on estrogen as teenage years to the menopause; its complex, chronic,

1048 https://academic.oup.com/edrv doi: 10.1210/er.2018-00242


REVIEW

ESSENTIAL POINTS
· Pelvic endometriosis, manifested by chronic pelvic pain and infertility, is a complex syndrome characterized by an
estrogen-dependent chronic inflammatory process that affects primarily pelvic tissues, including the ovaries, caused by
repeated retrograde travel and survival of shed endometrial tissue in the lower abdominal cavity
· The underlying pathologic mechanisms in the intracavitary endometrium and extrauterine endometriotic tissue involve
defectively programmed endometrial mesenchymal progenitor/stem cells
· Although endometriotic stromal cells, which compose the bulk of endometriotic lesions, do not carry somatic mutations,

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
they demonstrate specific epigenetic abnormalities that alter expression of key transcription factors such as excessive
production of GATA-binding factor-6, steroidogenic factor-1, and estrogen receptor-b, which collectively cause estrogen-
dependent inflammation, and deficient expression of progesterone receptor, which causes progesterone resistance
· Populations of endometrial and endometriotic epithelial cells harbor multiple cancer driver mutations, such as KRAS,
which may be associated with the establishment of pelvic endometriosis or ovarian cancer
· It is not known how interactions between epigenetically defective stromal cells and the mutated genes in epithelial cells
contribute to the pathogenesis of endometriosis
· Endometriosis-associated pelvic pain is currently managed by suppression of ovulatory menses and estrogen production,
cyclooxygenase inhibitors, and surgical removal of pelvic lesions, whereas in vitro fertilization is frequently used to
overcome infertility
· Although novel targeted treatments are becoming available, as endometriosis pathophysiology is better understood,
simple preventive approaches such as long-term ovulation suppression are currently underused

and systemic nature; the variety of tissues involved, uninterrupted and repetitious episodes of ovulation
including the central nervous system; and the need for giving rise to heavy periods associated with retrograde
treatments that address long-term suppression of menstruation, and pelvic endometriosis visualized by
ovulation (, ). laparoscopy are linked and likely represent progressive
Pelvic endometriosis, which may involve pelvic stages of a common disorder (Fig. ) (). (ii) De-
peritoneal surfaces, subperitoneal fat, rectovaginal bilitating menstrual pelvic pain first experienced
space, or ovaries, occurs primarily via retrograde during or soon after menarche gradually increases in
menstruation and comprises the vast majority of all severity as the inflammatory stimuli from pelvic dis-
cases of endometriosis (Fig. ). The disease may also ease persist and the peripheral and central nervous
affect the bladder, bowel (most commonly the rectum system are recurrently conditioned, leading to the
and appendix), deep pelvic nerves, ureters, anterior phenomenon of central sensitization (, ). Patients
abdominal wall, abdominal skin, diaphragm, pleura, with endometriosis experience chronic pain contin-
lungs, pericardium, and brain (). The symptoms of ually or intermittently until the menopause. (iii) All
pelvic endometriosis—painful periods, painful in- three forms of pelvic endometriosis, namely, perito-
tercourse, and chronic pelvic pain and infertility— neal endometriotic implants, rectovaginal nodules,
often disrupt the social, professional, academic, and and ovarian endometriomas, are intimately linked
economic potential of young women. Living with with ovulatory menses and probably originate pri-
severe cyclic or continuous pelvic pain or the threat of marily from retrograde menstruation that follow
its return, often for decades, can also lead to anxiety ovulation (Fig. ) (). (iv) The current diagnostic
and depression (). Another key source of stress standard, gross laparoscopic visualization of pelvic
associated with endometriosis is the potential com- endometriosis, does not take into account the presence
promise of current or future fertility (). Herein, we of microscopic inflammatory disease in the pelvic
review the clinical, biological, and genetic advances peritoneum or eutopic endometrial tissue, both of
that have been made in the area of endometriosis which may be the source of chronic pain that responds
during the past two decades, which may inform the to suppression of ovulation or estrogen formation (,
development of treatment and prevention approaches ). Thus, endometriosis should be a clinical diagnosis
for this debilitating disease. of exclusion and based on a detailed history and
symptoms, as clinicians in practice primarily manage
Salient medical features the symptoms of the disease. (v) Treatment-naive pa-
Taking a broad perspective of endometriosis—from tients with endometriosis significantly benefit from
the molecular mechanisms to its impact on quality of surgical or medical management, which leads to sub-
life—is essential for optimizing management that will stantial pain relief for practically all of these patients
benefit patients suffering from this disease (). The (); however, the response rate progressively and
following are considered key features of endometriosis. sharply decreases with each treatment attempt (, ).
(i) Intense primary dysmenorrhea (painful periods), Therefore, management of recurrent disease or its

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1049


REVIEW

symptoms is the key clinical challenge in endome- T cell expressed and secreted (RANTES), and
triosis (). (vi) Long-term suppression of ovulatory monocyte chemoattractant protein- (MCP-) (,
menses is the mainstay of any successful manage- , ). (iv) The importance of estrogen excess in
ment strategy for endometriosis-associated pelvic endometriosis is somewhat similar to the role of
pain (, ). (vii) Epidemiologic studies have found insulin deficiency in diabetes or the metabolic
significant associations between the prevalence of syndrome (–). Estradiol is essential for endo-
endometriosis and ovarian cancer (). The direct metrial tissue attachment to peritoneum; lesion
observation of malignant transformation of endo- survival; production of inflammatory substances

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
metriosis to an adenocarcinoma, however, has been such as metalloproteinases, cytokines, or prosta-
reported only rarely (). Currently, the potential for glandins and growth factors; and angiogenesis (Fig.
malignant transformation of endometriosis is not ) (–). Estrogen receptor (ER)b mediates the
considered a significant factor in standard clinical effects of estradiol in endometriosis and triggers
decision-making, which should be revisited and pathways that enhance lesion survival, remodel
updated in light of recently published data (, ). pelvic peritoneal tissue, and produce inflammatory
substances, which stimulate nociceptors in pelvic
Clinically useful research findings tissues, leading to pain (Fig. ) (, ). Blocking
The following key observations and cellular and estradiol production either naturally (menopause) or
molecular characteristics of endometriosis are pharmacologically (ovarian suppression or aroma-
helpful starting points for interpreting more in- tase inhibition) causes regression of the disease and
depth and complex mechanistic studies of the dis- its symptoms, including pain (Fig. ) (, , ). (v)
ease. (i) Menstruation that follows an ovulatory cycle Endometriosis is a unique condition whereby pro-
is indispensably linked to the development of en- gesterone resistance occurs owing to a deficiency of
dometriosis, at least in its early stages (Fig. ). Species progesterone receptor (PR) in endometriotic stromal
that do not menstruate do not develop endome- cells (Fig. ) (–). Endometriotic stromal cells
triosis, and women who never had ovulatory menses produce large amounts of progesterone locally; this
do not develop endometriosis. Interruption of is consistent with progesterone resistance resulting
ovulatory menses in women with endometriosis is from PR deficiency (). This characteristic also
usually therapeutic (Fig. ) (, ). (ii) Pathology in explains, in part, why endometriotic tissue responds
various tissue types may contribute to the overall poorly to progesterone or its agonists. Alternatively,
clinical picture of endometriosis. Distinct cellular synthetic antiprogestin compounds such as mifepristone
and molecular abnormalities involving inflam- and ulipristal acetate (UPA), which bind to low levels of
mation and steroid responsiveness have been well PR with higher affinity, suppress endometriosis and
described at least in two types of tissues: eutopic reduce pain more effectively (–).
(intrauterine) endometrium and ectopic endo- Recently, three groups independently described
metriotic tissue (). It is also plausible that defects in mutations affecting PIKCA, KRAS, ARIDA, and
uterine spiral arteries and arterioles or pelvic peri- other cancer driver genes in the epithelial components
toneal tissue may predispose certain women to of benign ovarian and extraovarian pelvic endome-
endometriosis (, ). The list of affected tissues triosis tissue (, , ). Stunningly, the epithelial cells
may grow as we understand more about endome- of clinically and histologically normal endometrial
triosis (, ). It follows that surgical removal of samples also contained mutations in mutant allele
pelvic endometriotic tissue, even by the most capable frequencies similar to those observed in endometriotic
surgeons, will not be curative. (iii) Most endo- epithelium and some cancer types (, ). These well-
metriotic implants are composed primarily of characterized driver mutations have also been reported
stromal cells and contain a small epithelial com- in clear cell and endometrioid-type ovarian cancers
ponent that lacks the deep invaginations (glands) uniquely associated with endometriosis, as well as in
typically observed in the eutopic endometrium (Fig. other epithelial ovarian adenocarcinomas (, ). It
). The endometriotic stromal cell is epigenetically appears that fragments of inflamed endometriotic
misprogrammed and displays partial phenotypes of stroma, together with mutated epithelium, may lo-
ovarian theca/granulosa cells and tissue macro- calize into ovarian inclusion cysts and transform to
phages. For example, endometriotic stromal cells cancer, possibly because the unique microenviron-
express the full cascade of steroidogenic proteins ment within the ovary becomes permissive to carci-
and enzymes such as steroidogenic acute regula- nogenesis (, ). Although the same phenomena
tory protein (STAR) and aromatase and convert and mutations are observed in extraovarian deep-
the precursor molecule cholesterol to substantial infiltrating endometriosis, the risk of malignancy in
quantities of progesterone and estradiol (). The such sites is negligible, as the extraovarian location is
endometriotic stromal cell also expresses and se- probably not supportive of malignant transformation
cretes large amounts of immune molecules such as (). The significance of these epithelial cancer driver
IL-b, IL-, TNF, regulated upon activation normal mutations in histologically normal-appearing and

1050 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

clinically disease-free eutopic endometrial tissue re- cells, and (iii) signs of chronic bleeding in or adjacent
mains enigmatic. to endometrium-like tissue, including collections of
red blood cells and hemosiderin-laden macrophages
Prevalence and distribution of endometriosis (immune cells that engulf blood pigment, Fig. ).
Clinically, it is more meaningful to discuss the Fibrosis comprised of fibroblasts and extracellular
prevalence of endometriosis rather than its incidence matrix is commonly observed surrounding endo-
because an average patient may suffer from the disease metriotic implants and possibly represents extensive
for prolonged periods of time—up to decades (). The inflammation and tissue remodeling (Fig. ). From a

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
disease process can begin as early as at the time of first surgical perspective, there are three major forms of
menses, and it may persist until after the menopause pelvic endometriosis: (i) peritoneal endometriosis
(, ). This natural history expands the pool of found on uterine serosa or peritoneal or subperitoneal
patients affected by endometriosis at any given time. tissue in pelvic side walls (Fig. a), (ii) deep infiltrating
An estimated  in  reproductive-age women—or
~ million women worldwide—may be suffering
from endometriosis (). In women undergoing ab- (a) Lower uterus
dominal surgery for chronic pelvic pain or infertility, Cervix
endometriosis represents one of the most common
pelvic pathologies (, ).
The most significant risk factor and cellu- Blood clot establishing endometriosis?
lar mechanism underlying pelvic endometriosis is
retrograde menstruation (Fig. ) (–). The risk of Right pelvic side wall
endometriosis appears to increase with a greater
lifetime number of ovulatory cycles (Fig. ) (). The Right tube
gradual but consistent trend toward more menstru- Free blood and endometrial and adnexa Right
ations due to earlier ages of menarche and a pro- tissue fragments in the ovary
retrovaginal pouch
gressively decreasing number of pregnancies per
woman (which naturally disrupt ovulatory cycles) has Bowel
increased the risk of implantation of retrograde-
traveled endometrium on pelvic tissues (–). Re-
peated cycles of ovulation and the genesis of ovarian (b) Retrograde menstruation (c) Peritoneal endometriosis
endometriosis have been linked, as endometriomas
RV pouch
seem to develop from ovarian follicles () and by
direct transition from hemorrhagic corpus luteum
cysts, based on serial ultrasound examinations (Figs. 
and ) (). It is quite likely that retrograde menstrual
endometrial material trapped in a recently ruptured
ovarian follicle gives rise to ovarian endometriomas
(). It was also suggested that endometrial fragments
on the ovarian surface epithelium may get trapped in
cortical invagination cysts and give rise to endome-
triomas; this possibility, however, was solely based on
histopathological examinations but not on temporal Epithelial cell
evidence as in the case of the ovarian follicle hy- Stromal cell

pothesis (, ).


(d) Deep-infiltrating endometriosis (e) Ovarian endometrioma
Histopathology
As discussed above, the term endometriosis initially
referred to a histologic diagnosis, but as the clinical
picture has become clearer with recent clinical and
molecular discoveries, the definition has evolved to
describe a symptom complex that affects the pelvic
tissues. Anatomically, pelvic endometriosis refers to
the presence of endometrium-like tissue outside of the
uterine cavity, usually on the pelvic peritoneal surfaces,
in the ovary or on other pelvic tissues such as bowel
(Fig. ). Surgical pathologic diagnosis is made upon the
identification of at least two of three key elements: (i)
endometrial stromal cells, (ii) endometrial epithelial

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1051


REVIEW

endometriosis that involves substantial fibrosis and endometrioma, an ovarian cyst with a wall that is lined
surrounding tissue remodeling and is usually found in up with endometrial tissue and contains substantial
the pouch between the vagina and the rectum, that amounts of clotted and unclotted blood products in its
is, rectovaginal nodule (Fig. b), and (iii) ovarian lumen (Fig. c–e).

Mechanisms of Endometriosis Moreover, most molecular and clinical data accumu-


lated during the past two decades support Sampson’s

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
Tissues that contribute to endometriosis postulate as the main mechanism for pelvic endometriosis
Several mechanisms have been proposed regarding the (, ). Thus, this review centers on Sampson’s ret-
histologic origins of endometriosis. Sampson () and rograde menstruation hypothesis and considers the
others postulated that fragments of menstrual endo- eutopic endometrium as the key tissue source for the
metrium pass retrograde through the fallopian tubes, genesis of pelvic endometriosis.
then implant and persist on peritoneal surfaces (Fig. ).
This has been demonstrated in primate models and
observed naturally in humans; it is also supported Intrauterine (eutopic) endometrium
by the observation that spontaneous endometriosis Retrograde flow followed by implantation of in-
occurs exclusively in species that menstruate, that trauterine endometrial tissue is the most plausible
is, female primates, including humans (). Young mechanism for most pelvic endometriotic lesions (Fig.
women with a transverse vaginal septum or imper- ) (). Reflux menstruation of endometrial tissue
forate hymen that blocks the expulsion of menstrual mixed with blood is routinely observed in almost all
tissue from the vagina almost invariably develop pelvic ovulatory women, but only ~% of these women
endometriosis (). An alternative hypothesis is that the develop the symptom complex of endometriosis.
peritoneum, derived from the coelomic epithelium, Several explanations have been offered for how
undergoes metaplasia to differentiate into islands of refluxed endometrium to the peritoneal surface suc-
endometriotic lesions within the peritoneal cavity (). cessfully implants and survives in the long term in a
The defenders of yet another hypothesis suggest that limited population of women (). (i) The mucosal
menstrual tissue from the endometrial cavity reaches surfaces of the uterus, that is, eutopic endometrium
pelvic or other distant body sites via veins or lymphatic and the tubal mucosa, in women with endometriosis
vessels (). Additionally, it has been proposed that display a number of cellular and molecular abnor-
circulating blood cells originating from bone marrow malities. Molecular defects have been reported in
differentiate into endometriotic tissue at various body eutopic endometrial tissues of women with endo-
sites (). Distant organ endometriosis such as lung metriosis, such as activation of oncogenic pathways or
endometriosis is very rarely encountered and may be biosynthetic cascades that favor increased production
explained by vascular spread. However, it is highly of estrogen, cytokines, prostaglandins, and metal-
challenging to construct clinically relevant models to loproteinases (Fig. ) (, , –). Upon the at-
support or refute the mechanisms proposed as alterna- tachment of these defective tissues to the peritoneum
tives to Sampson’s retrograde menstruation theory. or the ovary, the magnitude of these abnormalities is
amplified drastically to enhance their implantation
and longevity (). (ii) Compared with endometriosis-
free women, women with endometriosis may simply
have overwhelmingly higher numbers of molecularly
Figure 1. (a) Laparoscopy of the pelvis performed at the time of menstruation. Predictable cyclic unremarkable endometrial tissue, including somatic
ovulatory menses giving rise to repetitious episodes of retrograde travel of endometrial tissue and stem cells, that arrive at the pelvic abdominal cavity
blood into the dependent portions of the pelvic cavity is the main cause of pelvic endometriosis. during each menstrual episode (Fig. ). Several groups
Not all women who experience retrograde menstruation, however, develop endometriosis. This proposed that endometrial somatic stem cells may be
suggests that a number of differences between the patients with endometriosis and disease-free involved in the pathogenesis of premenarchal and
women may account for this condition. These include increased quantities of menstrual tissue that
adolescent endometriosis through retrograde neonatal
reach the abdominal cavity because of outflow track obstruction or deeper separation of the
functionalis layer from the basalis layer (see Fig. 6) and cellular and molecular defects in eutopic uterine bleeding due to maternal progesterone with-
endometrial or peritoneal tissues of women with endometriosis. (b) Graphic depiction of drawal at birth (–). These endometrial somatic
retrograde flow of endometrial tissue fragments made of spindly stromal and cuboidal epithelial stem cells would remain dormant beneath the peri-
cells. (c and d) Menstrual tissue fragments may survive and grow on peritoneal or subperitoneal toneum until increasing estradiol levels before men-
locations (peritoneal endometriosis) or may get deposited into the rectovaginal (RV) pouch during arche activate them to initiate clonal growths of
repetitious episodes of menstruation and remodel the neighboring vaginal, rectal, and cervical
ectopic endometrium in the peritoneal cavity ().
tissues via a chronic inflammatory process to give rise to a deep-infiltrating RV nodule. (e) The
endometrial tissue fragments may populate the exposed lining of a follicular or corpus luteum cyst
It has also been suggested that a defective immune
to eventually evolve into an endometrioma. [Adapted with permission from Bulun SE. system might fail to clear implants off the peritoneal
Endometriosis. In: Strauss J, Barbieri R, eds. Yen & Jaffe’s Reproductive Endocrinology. 8th ed. surface (, , ). One can further imagine that
Philadelphia, PA: Elsevier; 2019:609–642. Copyright © 2019 by Elsevier.] defective spiral arterioles may lead to hypermenorrhea

1052 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

Figure 2. Laparoscopic views of pelvic endometriosis. (a) A raised superficial endometriotic implant on bowel serosa [visceral
peritoneum]. (b) Deep-infiltrating endometriosis. A laparoscopic image sometimes described as “frozen pelvis” because of extensive
endometriosis and diffuse tissue remodeling causing dense adhesions between the ovary, bowel (rectum), and the uterine peritoneum.
White vesicular endometriotic lesions are visible in the delineated area that represents the upper tip of diffuse adhesions caused by
endometriosis. A challenging dissection into this plane will eventually take the surgeon into the previously existing rectovaginal (RV)
space now harboring a nodule composed of endometriotic tissue and surrounding fibrosis and allow the removal of this RV nodule [see
Fig. 4b]. (c) Enlarged left ovary because of a large endometrioma buried in the normal tissue [see (d) and Fig. 4c]. (d) Dissection into the
overly stretched normal white-tan ovarian cortical tissue. The fibrotic endometrioma cyst wall is grasped by a forceps. The suction
apparatus was inserted into the cyst lumen to remove the thick chocolaty fluid composed of blood products. The surgeon will develop a

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
plane between the normal ovarian tissue and cyst wall in an attempt to remove the cyst in its entirety.

or other menstrual abnormalities and contribute to a aromatase, cyclooxygenase (COX), and IL- mRNA
phenotype that favors endometriosis (). It is quite and protein and display readily detectable levels of
likely that a whole variety of cellular and molecular aromatase enzyme activity that catalyzes the con-
abnormalities in the eutopic endometrium or else- version of androgens to estrogens (Fig. ) (, , ,
where may lead to the common phenotype of en- ). Transcriptomic analysis of eutopic endometrium
dometriosis together with its symptom complex. from women with or without endometriosis con-
Cellular and molecular aberrations in eutopic firmed these earlier observations and found dysre-
endometrium. Eutopic endometrium of women with gulation of selected genes that contribute to implantation,
endometriosis looks identical to that of disease-free including embryonic attachment, embryo toxicity, im-
controls under routine microscopic examination via mune dysfunction, and apoptotic responses, as well as
hematoxylin and eosin staining. However, substantial genes that likely contribute to the pathogenesis of en-
clinical and molecular data support the existence of dometriosis, including aromatase, PR, and angiogenic
cellular and molecular differences in eutopic endome- factors (Fig. ) ().
trial tissue of patients with endometriosis (Fig. ) (). Nerve tissue has been immunohistochemically
Stromal cells of eutopic endometrium of women identified in the functional layer of eutopic endo-
with endometriosis express strikingly higher levels of metrial tissue in all women with endometriosis but not

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1053


REVIEW

in the eutopic endometrium of disease-free women potential for mesodermal, endodermal, and ectoder-
(Fig. ) (). The pathologic presence of nerve fibers mal lineages, indicating their plasticity and multi-
points to a general defect in the differentiation of potency (). A recent study showed that, in contrast to
somatic stem cells in the endometrium of patients with endometrial mesenchymal stem cells from disease-free
endometriosis. This may have important implications patients, eutopic endometrial mesenchymal stem cells
for understanding the generation of pain in these isolated from patients with endometriosis did not
patients. For example, this may explain why patients differentiate in vitro to stromal cells that decidualize
with endometriosis continue to experience pain after properly (). This suggested that the observed pro-

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
surgical resection of ectopic implants (). These nerve inflammatory and progesterone-resistant gene ex-
fibers may be directly involved in transmitting pain pression signature in eutopic endometrial stromal
both in eutopic or ectopic locations (). tissue from patients with endometriosis originates
from defective endometrial mesenchymal stem cells
Progenitor/stem cells (Figs.  and ). Immunohistochemical analyses of
In ovulatory women, the progenitor/stem cell pop- endometrial tissues of patients undergoing bone mar-
ulations in the basalis layer of endometrium re- row transplants has also suggested that bone marrow
generates the full thickness mucosa every month cells may contribute small populations of endometrial
within days under the influence of estrogen (Fig. ). cell types (). It is conceivable that these circulating
Thus, stem cell activity is extraordinarily important for stem cells may be involved in rare cases of extraper-
endometrial function. Mesenchymal (stromal) stem itoneal lesions ().
cells isolated from endometrial tissue have similar The discovery of markers for endometrial stem/
properties to bone marrow mesenchymal stem cells progenitor cells has enabled the examination of their
(). Selective markers for mesenchymal stem cell role in endometriosis, adenomyosis, and healthy en-
enrichment, CD, b-type platelet-derived growth dometrial development for implantation (). How-
factor receptor (PDGFRB), and sushi domain ever, we are still far from establishing clinically relevant
containing- (SUSD), reveal a perivascular location of experimental models of human endometrial stem cells
mesenchymal stem cells and suggest a pericyte identity that regenerate endometrial tissue in response to es-
of these cells in blood vessels of the endometrium (). trogen as during the menstrual cycle (). Together
Molecular analyses of SUSD-positive cells demon- with the DNA methylation defects observed in eutopic
strate their phenotype as perivascular cells. Endome- endometrium of patients with endometriosis, these
trial stromal cells or stromal cells isolated from findings substantiate the extraordinarily important
menstrual blood also display mesenchymal stem cell and interactive roles of epigenomic mechanisms and
characteristics and demonstrate broad differentiation stem cells in the pathophysiology of endometriosis
(Figs.  and ) ().

Uterine vasculature
Endometrial vasculature, in particular, the spiral
arterioles play a critical role in the process of
menstruation (). Upon the withdrawal of pro-
gesterone and estrogen during the late luteal phase
of a nonpregnant woman, these vessels undergo
acute vasoconstriction and intravascular coagulation
accompanied by inflammatory cell infiltration and
degradation of extracellular matrix in adjacent en-
dometrial tissue. This leads to shedding and expul-
sion of the top endometrial layer named functionalis
along with blood. It was proposed that the spiral
arteries and arterioles may be molecularly different in
the uteri of women with endometriosis compared
with those in disease-free women (Fig. ) (). Owing
to these differences, it is plausible that the process of
separation of the functionalis layer from the basalis
layer may occur at a deeper plane in women with
endometriosis and heavy menses (Fig. ). Because
endometrial somatic stem cells may likely be con-
Figure 3. The indispensable roles of ovulation and estrogen in endometriosis, highlighting the
mechanisms of menstruation, tissue survival, inflammation and pain, and the targets of treatment
centrated in the deeper (basalis) portion of this tissue,
in endometriosis. E2, estradiol; P, progesterone. [Adapted with permission from Bulun SE. this may lead to an higher influx of stem-like cells
Endometriosis. In: Strauss J, Barbieri R, eds. Yen & Jaffe’s Reproductive Endocrinology. 8th ed. into the lower abdominal cavity via retrograde
Philadelphia, PA: Elsevier; 2019:609–642. Copyright © 2019 by Elsevier.] menstruation in women with endometriosis.

1054 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

Figure 4. (a) Peritoneal endometriosis with fibrosis. (b) Rectovaginal nodule with extensive fibrosis and tissue remodeling surrounding
islands of endometriotic stroma and occasional epithelial cells. (c) Sections of an ovarian endometrioma cyst. Note that the thickness of
the endometrial lining (stroma and luminal epithelium) varies throughout the cyst wall, with foci of bleeding and macrophages
containing blood pigment. The cyst wall is primarily composed of fibrotic tissue. (d and e) Higher-magnification pictures showing details
of the ovarian endometrioma cyst wall from (c).

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019

Peritoneum attachment to the peritoneal surface and its eventual


Theoretically, the formation of peritoneal endome- establishment into the subperitoneal space through
triosis requires the initial adhesion of shed endometrial breaks in the mesothelial cell layer happens over a
cells to peritoneal and subperitoneal surfaces. From a short period of time (Fig. ) (). Thus, a mesothelial
mechanistic perspective, this involves the expression of surface with physical and temporal “windows” that
extracellular matrix adhesion molecules and their facilitate the implantation and survival of endometrial
coreceptors (). Circumstantial and experimental tissue fragments may be essential for the formation of
observations suggest that the process of implant peritoneal endometriosis.

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1055


REVIEW

In contrast to the implantation theory, the mechanistic molecular studies that employ endo-
metaplasia theory suggests that the coelomic epi- metrial or endometriotic stromal cells, which may
thelium pathologically differentiates into endometrium- create a scientific bias against the potential roles of
like tissue to give rise to peritoneal endometriosis other cell types. Until very recently, relatively little
(). The idea of metaplasia is plausible, because cells has been known about the endometriotic epithelial
from both the peritoneum and endometrium derive cell (, , ). Although no somatic mutations have
from a common embryologic precursor called the been found in endometriotic stromal cells (), a
coelomic epithelium. However, it is challenging to stunningly high number of somatic mutations in

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
demonstrate this potential mechanism experimen- cancer driver genes have been recently discovered in
tally. Although overwhelming experimental data that the epithelial cells of both clinically and histologically
support the implantation of retrograde menstrual normal eutopic endometrium as well as in benign
material have accumulated, this is not the case for the pelvic endometriotic lesions (, , ). The effects
metaplasia theory (). Even so, the pelvic peritoneum of these mutations on epithelial cell fate, the
and its mesothelial surface seem to contribute neighboring stromal cells, and endometriotic tissue
critically to the establishment of endometriosis as a whole remain to be determined. Somatic epi-
significantly (). thelial mutations in endometriosis are discussed in
greater detail below. The following biologic pro-
cesses pertain to endometriotic stromal and epi-
Key biological processes in endometriosis
thelial cells.
Cell fate in endometriotic tissue has been studied
primarily at the level of the endometriotic stromal
cell, which dominates the lesion in terms of quantity Reduced apoptosis
and exhibits most of the key and therapeutically Apoptosis is significantly decreased in endometriotic
targetable molecular abnormalities of endometriosis, stromal and epithelial cells compared with eutopic
such as estradiol production, progesterone re- endometrial tissues (, ). This may be linked to
sistance, cytokine production, and prostaglandin pathologic levels of local estradiol biosynthesis ().
production (Fig. ) (). Endometrial or endometri- ERb mediates the antiapoptotic effects of estradiol in
otic stromal cells can be maintained in primary endometriotic stromal cells ().
culture to study their biology, whereas this is more
challenging with epithelial cells (, ). This technical Defective differentiation
limitation has led to a disproportionate number of A critical observation in endometriosis is deficient
differentiation (decidualization) of the stromal cell (,
, ), which has been linked to progesterone re-
sistance (, ). Although the first molecular evi-
dence of progesterone resistance was observed in
the endometriotic epithelial cell, deficient stromal
decidualization was eventually found to be the primary
mechanism (, ). Epigenetically misprogrammed
mesenchymal stem cells in endometriosis also drives
deficient differentiation (, ).

Inflammation
Inflammation is the central process in endometriosis.
It leads to pain, remodeling of neighboring tissues,
fibrosis, adhesion formation, and infertility. The
production of cytokines and prostaglandins and in-
filtration of immune cells are some of the hallmarks of
inflammation (). The endometriotic stromal cell is
one of the major sources of cytokines and prosta-
glandins (, ). Recurrent episodes of bleeding and
an attempt by macrophages to remove blood pigments
also contribute to the inflammatory process and ad-
hesion formation (Fig. b). Inflammatory processes are
thought to be induced by estradiol and mediated by
Figure 5. Overview of the complex roles of retrograde menstruation, epigenetically defective ERb in endometriosis, as denial of ovarian or local
endometrial stromal cells, the epithelial cells carrying mutations, DNA methylation, nuclear estradiol to endometriotic tissue using a GnRH analog
receptors, and inflammation in endometriosis. E2, estradiol. Endometrial tissue fragments may and/or an aromatase inhibitor stops or decreases pain
implant on pelvic peritoneal tissue surfaces or may get trapped in an ovarian inclusion cyst such as
(Fig. ) (, , , , ).
a hemorrhagic corpus luteum cyst.

1056 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

Proliferation Figure 6. Possible interactions between spiral arterioles,


In eutopic endometrial tissue, the epithelial cell somatic stem cells, and menstruation and the risk of
proliferates under the influence of estrogen (). endometriosis. Menstruation occurs after the functionalis layer
of endometrium separates from the basalis and is expelled
The strikingly rapid rate of proliferation possibly con-
through the cervix or uterine tubes. This separation occurs in
tributes to the acquisition of somatic epithelial association with vasoconstriction and coagulation in the spiral
mutations in eutopic endometrial tissue (). Most arterioles giving rise to degradation of the extracellular matrix,
endometriotic implants, however, contain scant epithe- hypoxia, and necrosis in the separated segment. The basalis
lium and are comprised primarily of stromal cells (Fig. ). layer may contain higher numbers of functional progenitor cells

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
In contrast to an epithelial cell, human mesenchymal that will regenerate the functionalis layer during the next
proliferative phase under the influence of estrogen. It is
cells are not epigenetically programmed to proliferate
plausible to hypothesize that the separation between the two
extensively, nor are they prone to accumulate mutations. layers at a relatively superficial plane may lessen the likelihood
Therefore, endometrial or endometriotic stromal cells of heavy bleeding or travel of stem cells into the pelvic cavity.
show limited proliferative activity. In fact, significantly Separation at a deeper plane, however, may favor the
less proliferative activity was reported in endometriotic endometriosis phenotype. One can envision, therefore, that
tissue compared with eutopic endometrial tissue (). possible epigenetic abnormalities in the vascular system may
One exception is in abdominal wall endometriosis. affect the process of separation between the functionalis and
basalis and thus affect the risk for endometriosis. The somatic
Endometriotic lesions seem to grow quite extensively in
stem cells per se may also be defective, or simply larger numbers
the subcutaneous adipose tissue in the abdominal wall of otherwise normal stem cells that travel into the abdominal
(), likely driven by significant proliferative activity. It is cavity may increase the risk for endometriosis. (Adapted with
tempting to hypothesize that aromatase activity in adi- permission from Bulun SE. Endometriosis. In: Strauss J, Barbieri
pose fibroblasts in subcutaneous fat tissue may con- R, eds. Yen & Jaffe’s Reproductive Endocrinology. 8th ed.
tribute to this process (). Importantly, note that Philadelphia, PA: Elsevier; 2019:609–642. Copyright © 2019 by
aromatase activity in human subcutaneous adipose tissue Elsevier.)
is significantly higher than that in the omentum or
visceral (e.g., subperitoneal) adipose tissue ().

Angiogenesis
Formation of blood vessels and their steroid
hormone–dependent function are routinely observed
in endometriotic lesions. In fact, recurrent and chronic
bleeding evident by hemosiderin-laden macrophages
is a unique histologic feature of endometriosis (Fig. ).
Cytokines such as IL-A may enhance angiogenesis
in the peritoneal cavity for the establishment and
growth of endometriotic lesions (). Moreover, an
ERb-selective estradiol agonist stimulated the ex-
pression of SLIT that may play a key role in en- endometrial cancer) or benign (e.g., uterine leio-
hancing angiogenesis in endometriosis (). myoma) neoplasms (). Estrogen-driven inflamma-
tion seems to be the central process that shapes the
Tissue remodeling pathology of endometriosis.
Some survival capability seems to be necessary for the
initial attachment of endometrial tissue fragments to Genomic alterations
pelvic peritoneum and the establishment of sub- Genomic alterations affect the structure and function of
peritoneal endometriotic lesions (). Once the initial the complete set of DNA including all of its genes and
implantation of the endometrial tissue fragment oc- noncoding intergenic regions. Genomic mechanisms
curs, limited proliferation and tissue growth may be affect the regulation of all genes, which direct the
necessary for the long-term survival of the tissue. production of proteins with the assistance of epi-
Under the influence of estrogen, inflammation ensues genomic modifications (see below). Proteins make up
and causes significant remodeling of the peritoneal cells and tissues, regulate chemical reactions, and carry
and subperitoneal tissues, such as adipose tissue (Fig. signals between cells. Recent advances in genomics have
B and Fig. ). One of the best studied pathways that revolutionized how we approach complex systems in
mediates surrounding tissue remodeling involves high health and disease. Intensive efforts have been made to
metalloproteinase activity in endometriotic implants understand genome-wide mechanisms in endometri-
(). In fact, fibrosis of the surrounding tissue is a osis. Selected studies are highlighted below.
hallmark of peritoneal or ovarian endometriosis (Fig.
) (). Invasion and proliferation, however, are Inheritance
much less important to the pathology of endometriosis Endometriosis as a phenotype seems to be transmitted
than they are to the pathology of malignant (e.g., in families in a polygenic manner (). In one study,

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1057


REVIEW

the mothers and sisters of women with severe en- in a subanalysis of  families with more than three
dometriosis had a sevenfold higher likelihood of affected members, suggesting that the presence of
suffering from endometriosis compared with primary one or more rare variants confers high risk; however,
female relatives of their partners (). Consistent with no mutations could be identified (, ). To
this observation is the finding that familial cases of date, family-based or case-control genetic association
endometriosis tend to be more severe and have an studies have not led to any biologically meaningful
earlier onset of symptoms than do sporadic cases results.
(). Based on a study of  twins, the heritability

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
of endometriosis, or the proportion of disease variance Genome-wide association studies in endometriosis
due to genetic factors, has been estimated at ~% Genome-wide association studies (GWASs) typically
(). These studies of familial endometriosis point to focus on associations between single-nucleotide
genetic transmission of the disease. The mode of polymorphisms (SNPs) and traits such as major hu-
transmission, however, seems to be complex, as in man diseases and do not require the use of pedigrees.
other chronic inflammatory conditions such as in- Since , a number of widely acknowledged en-
flammatory bowel disease. dometriosis GWASs have been published; these
studies employed data sets involving thousands of
Candidate gene studies endometriosis cases and controls, primarily women of
Since the s, numerous large-scale international Japanese or of European ancestry (, , ).
studies have tried to identify candidate genes that lead Meta-analyses of these GWAS data sets confirmed
to the endometriosis phenotype. A number of “off-the- their findings and revealed additional loci (). Many
shelf” candidate gene loci have been interrogated based GWAS-identified loci were reported in women with
on their biological plausibility and possible connection endometriosis. A subset of these is discussed below.
with endometriosis, but these studies were negative Endometriosis GWASs collectively provided sig-
(, ). Case-control genetic association studies, nificant evidence for association of loci near:
performed primarily to find polymorphisms, also cyclin-dependent kinase inhibitor B antisense RNA
failed to discover any function-altering genetic variants (CDKNB-AS), an intergenic region on chromosome
in familial endometriosis (–). Geneticists have p., wingless-type MMTV integration site family
cited the following factors for the failure of candidate member- (WNT), vezatin (VEZT), an intergenic
gene studies to ascertain the genetic basis of complex region on p, and inhibitor of DNA binding- (ID)
inflammatory diseases such as endometriosis. (i) The (). Some of these loci had stronger effect sizes among
underlying biological hypothesis may not be valid. (ii) American Society of Reproductive Medicine advanced
These studies typically investigate a limited number stage (III to IV) cases, implying that they are likely to be
of genes in a potentially important biological path- implicated in the development of moderate to severe, or
way. (iii) A limited number of variants in a gene ovarian, disease (). Results from follow-up papers
are tested. (iv) Cases (endometriosis) and controls have either confirmed or questioned these findings and
(endometriosis-free) may not be optimally defined. (v) highlight the heterogeneity in the genetic loci that
Sample sizes may not be sufficient to detect the effect underlie endometriosis in different populations, even
sizes that are expected for variants influencing a when sampled from women with similar ethnic ancestry
complex trait (). The key reason for the lack of (). Despite these large-scale and expensive studies,
meaningful results, however, could be as simple as the no mutations affecting these genes in familial cases of
actual absence of any classically defined germ cell endometriosis were reported. To date, these studies
mutations that cause the endometriosis phenotype. have not resulted in the identification of any thera-
peutically targetable molecules or gene products.
Genetic linkage analysis Meta-analyses of endometriosis GWASs also
Using genetic linkage analysis, an international study identified loci containing genes involved in ovarian
group analyzed  affected sister-pair families steroid hormone pathways: growth regulation by es-
and found evidence of significant linkage of endo- trogen in breast cancer- (GREB), coiled-coil domain
metriosis to chromosome q (). Subsequent containing  (CCDC), ERa (ESR), spectrin
fine-mapping association analyses of chromosome repeat containing, nuclear envelope- (SYNE), and
q suggested a possible association of common FSH b-subunit (FSHB) (, ). The roles of FSHB
variants near the CYPC gene, but mutations that and ERa in ovarian function and estrogen action
could explain the linkage signal could not be dem- are well known (). The product of the estradiol/
onstrated (, ). Note that CYPC encodes ERa target gene GREB increases proliferation and
an important drug-metabolizing enzyme involved extracellular matrix formation by ovarian cancer cells
in the biotransformation of proton pump inhibitors (). GREB is expressed in % to % of serous,
and antidepressants (). Estradiol via ERa inhibits endometrioid, mucinous, and clear cell carcinomas.
CYPC expression (). The same group later Serous, endometrioid, and mucinous ovarian cancers
identified significant linkage to chromosome p– are almost always positive for either ESR or GREB,

1058 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

suggesting a possible reliance on signaling through endometria (Fig. ) (). In endometriotic and normal
ESR and/or GREB (). In breast cancer, recur- endometrial epithelial cells, numerous somatic mu-
rent rearrangements are seen between ESR and its tations were identified within genes frequently mu-
neighbor CCDC, which are enriched in the more tated in endometriosis-associated epithelial ovarian
aggressive and endocrine-resistant luminal-B tumors cancers. PIKCA and KRAS were found to be the most
(). GWASs also demonstrated a critical missense frequently mutated genes in ovarian endometriotic
SNP located  kb downstream of ESR in another epithelial cells (Fig. ). In particular, there was a sig-
neighboring gene, SYNE (). SYNE is involved in nificantly higher mutant allele frequency for KRAS

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
nuclear organization and structural integrity, function (Fig. ) (). An exception was ARIDA, a gene fre-
of the Golgi apparatus, and cytokinesis (). An iso- quently mutated in ovarian clear cell cancer, a ma-
form encoded by SYNE is downregulated in ovarian lignancy epidemiologically associated with pelvic
and other cancers; this SNP was genotyped in patients endometriosis (Fig. ) (). Although ARIDA muta-
with serous and mucinous ovarian cancers (). tions were frequently detected in endometriotic epi-
Some of these loci have stronger effect sizes among thelium, they were absent or very rare in endometrial
advanced stage (III to IV) endometriosis, implying that epithelium (). A detailed sequencing analysis of .
they are likely to be implicated in the development of single intrauterine endometrial glands (deeply in-
moderate to severe ovarian disease (). Despite these vaginated epithelial cells) demonstrated that each gland
large-scale and expensive genetic linkage studies or carried distinct cancer-associated mutations, suggesting
GWASs, no germline mutations affecting these genes in the heterogeneity of the genomic architecture of en-
familial cases of endometriosis have been reported. In dometrial epithelial cells (). It was proposed that
summary, endometriosis GWASs identified genes as- significant increases in mutant allele frequency in
sociated with uterine development and stem cell cancer-associated genes in endometriotic epithelium are
function (WNT), ovulatory function (FSHB, ESR), consistent with retrograde flow of endometrial cells
and estrogen action (ESR, GREB, CCDC, SYNE, already harboring cancer-associated mutations. If these
CYPC) (, , ); notably, most of these genes tissue fragments implant at ectopic anatomic locations
also have associations with breast or ovarian cancer with selective survival-supporting advantages, this may
(–). As the mechanistic link between endome- increase the risk for the development of endometriosis
“Despite these large-scale and
triosis and ovarian cancer becomes more clear, the roles and also malignant transformation (Figs.  and ) ().
expensive studies, no
of these genes with SNPs may become more apparent. It follows that the ovary, particularly a hemorrhagic mutations affecting these
corpus luteum cyst, is fertile ground for the development genes in familial cases of
Somatic epithelial mutations in endometrium of endometriosis and its malignant transformation. The endometriosis were reported.”
and endometriosis ovarian microenvironment, with massive levels of es-
Recently, a number of exome-wide sequencing studies trogen and possibly other mitogens of cancer cells, may
reported somatic epithelial mutations in ovarian contribute to this phenomenon.
endometriomas and nonovarian deep-infiltrating en- Another recent American exome-sequencing
dometriosis as well as in eutopic endometrium (Figs.  study, published in , targeted extraovarian
and ) (, , ). In , a group of investigators from deep-infiltrating endometriotic tissue and examined
China was the first to report a large number (thousands) both epithelial and stromal components of endo-
of single nucleotide variants in laser-microdissected metriotic tissues (). No mutations were detected in
epithelial cells from ovarian endometriosis and endometriotic or endometrial stromal cells (Fig. )
matched eutopic endometrial epithelial cells and in (). Whole-exome sequencing of epithelial cells of
endometrial epithelium from endometriosis-free women extraovarian endometriosis demonstrated somatic
(). Most nonsynonymous and synonymous single- mutations in ~% of patients (Fig. ). The mutant
nucleotide variants were commonly present in both allele frequencies of somatic mutations were quite low,
ectopic and matched eutopic endometrial cells and however, at ,%. This suggests that that only a
associated with genes involved in chromatin remodeling subgroup of endometriotic epithelial cells had mu-
and cell adhesion (). These authors did not particularly tations (). Although % of endometriotic tissues
emphasize any ovarian cancer driver genes, but they carried known ovarian cancer driver mutations in
focused on the possible patterns of mutagenesis at the ARIDA, PIKCA, KRAS, or PPPRA, it was highly
base pair level (). Surprisingly, they observed a large improbable that these driver mutations occurred at
number of nonsynonymous single-nucleotide variants this rate incidentally (P 5 .; Fig. ) (). A KRAS
predicted to alter protein structure in endometrial mutation was detected in % to % of epithelial
epithelial cells from endometriosis-free controls (Fig. cells in each endometriotic lesion, as shown by
) (). The numbers of mutations were comparable to droplet digital PCR analysis (). KRAS poly-
those found in common human malignancies (). morphisms reported in patients with endome-
A Japanese study published in  examined laser- triosis further support a role of this gene ().
microdissected cells from . ovarian endome- Because the risk of cancer in extraovarian endome-
triomas and a large number of control eutopic triosis is negligible, the clinical significance of these

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1059


REVIEW

Figure 7. Roles of endometriotic cell types with distinct abnormalities. Endometriotic stromal cells do not contain nonsynonymous
base pair alterations (mutations) but display extensive epigenetic defects that regulate the expression or silencing of genes. Specific
patterns of DNA methylation (C) or demethylation (s) give rise to the suppression or overproduction of specific proteins. In
endometriotic stromal cells, GATA6, ERb, and SF1 proteins are overproduced, whereas PR is suppressed, leading to progesterone
resistance. These changes collectively cause the accumulation of inflammatory and tissue-remodeling substances, including PGE2, E2,
cytokines, and matrix metalloproteinases (MMPs). In contrast, portions of the eutopic endometrial or endometriotic epithelial cells
accumulate tumor driver mutations that disrupt or change the function of critical proteins, including Kirsten rat sarcoma homolog
(KRAS), phosphatidylinositol-3-kinase, catalytic a polypeptide (PIK3CA), AT-rich interactive domain-containing protein 1A (ARID1A),
and numerous other oncogenes or tumor suppressors. Extraordinarily high concentrations of E2 and its metabolites in the ovary and

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
stroma-derived inflammation may contribute to the accumulation of epithelial mutations. [Chronic inflammation is suspected to cause
mutagenesis and carcinogenesis in other tissues such as Barrett’s esophagus and esophageal cancer (208).] The relative contributions of
endometriotic stromal vs epithelial cells to the development of pelvic endometriosis are currently unknown. The effects of each cell
type on the acquisition of genome-wide epigenetic defects or mutagenesis are also not known.

sporadic epithelial mutations in deep-infiltrating en- as opposed to genomic or genetic alterations that
dometriosis remains to be determined (). Similar involve base pair changes that may change the
driver mutations in the epithelial cells of ovarian structure and function of a protein encoded by a
endometriomas, however, may play key roles in ini- particular gene (Fig. ). Two of the most characterized
tiating clear cell, endometrioid cell, or possibly other epigenetic (single genes) or epigenomic (genome-
types of ovarian adenocarcinomas (Fig. ) (). Ad- wide) modifications are DNA (cytosine) methylation
ditionally, the epithelial mutations in extraovarian and histone modification (methylation or acetylation
deep-infiltrating endometriosis may play critical roles of specific histones in the chromatin). Studies of
in the implantation and establishment of endo- endometriotic stromal cells provided clues about
metriotic lesions in these locations, although they may epigenomic changes that lead to inheritable traits,
not become malignant. without altering the DNA sequence (Fig. ). Some of
these studies are reviewed below and link epigenetic
Epigenomic alterations modifications to functional consequences such as
Epigenomic mechanisms affect all genes and inter- steroid production and action.
genic regions in DNA, which are packaged together
with proteins (e.g., histones) into a structure known as Genome-wide defective DNA programming in
chromatin. Epigenetic modifications are reversible endometriotic and endometrial stromal cells
alterations of a cell’s DNA or histones that regulate Unlike endometriotic epithelial cells, endometriotic
gene expression without altering the DNA sequence, stromal cells do not carry any somatic mutations, but

1060 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

the expression levels of many genes in endometriotic selected to survive as endometriotic implants in the
stromal cells are strikingly different than those in pelvis (Fig. ).
eutopic endometrial stromal cells of women with or
without endometriosis (, ). This prompted in- GATA-binding factor-2/GATA-binding factor-6:
vestigators to examine potential epigenomic alter- opposing master regulators of physiology
ations in endometriosis. Although genome-wide and pathology
alterations in histone modifications have not been well Differential DNA methylation and expres-
defined in endometriosis, differential DNA methyla- sion. Numerous CpGs throughout the promoter and

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
tion in single genes as well as genome-wide has been coding region of GATA-binding factor- (GATA)
studied extensively (Fig. ) (). showed higher methylation in endometriotic stromal
Methylation of DNA is initiated and maintained by cells relative to endometrial stromal cells, whereas
three DNA methyltransferases (DNMTs)—DNMT, GATA-binding factor- (GATA) had less methyla-
DNMTA, and DNMTB—which catalyze the addi- tion across ranges of intronic CpGs flanking CpG
tion of methyl groups to the 9 carbon of cytosine in islands in endometriotic stromal cells (Fig. ) ().
targeted cytosine-phosphate-guanine dinucleotides Detailed mapping showed complete unmethylation of
(CpGs) (). Among these, DNMTB seems to be the GATA exon  in endometrium; this region was
differentially expressed in eutopic endometrial and predominantly methylated in endometriosis (Fig. )
endometriotic stromal cells during the decidualization (). Likewise, exon  of GATA showed full meth-
process (). Additionally, DNMTB also binds to ylation in endometrium and full unmethylation in
the promoter regions of key genes, steroidogenic endometriosis (Fig. ) (). GATA protein was highly
factor- (SF) and ESR, in endometrial vs endo- abundant in endometrial stromal cells with a strong
metriotic stromal cells (). Thus, differential nuclear signal, but it was scarcely detectable in
DNMTB expression and binding to critical gene endometriotic stromal cells. In contrast, GATA was
promoters in endometriotic stromal cells may con- robustly expressed and localized to the nuclei in all
tribute to an aberrant DNA methylome that misdirects endometriotic stromal cells but barely detectable in
gene expression in endometriosis and contributes to endometrial stromal cells ().
these cells’ altered response to steroid hormones (). Physiologic functions of GATA2 in endo-
Genome-wide differences in DNA methylation metrium. In mice, conditional GATA depletion in
between endometriotic and normal endometrial uterine tissue resulted in diminished PR expression in
stromal cells have been reported and were correlated uterine epithelial cells and attenuated progesterone
with gene expression using an interaction analysis signaling. Moreover, GATA depletion in mouse
strategy (). Out of some , methylation uterine tissue gave rise to infertility due to implan-
sites, ., differentially methylated CpGs were tation failure (). In human endometrial stromal
identified in endometriotic stromal cells compared cells, however, GATA depletion did not alter PR,
with normal endometrial stromal cells (Fig. ) (). ERa, or ERb levels (), or the expression of some
Differential CpG methylation patterns were seen most other critical endometriosis-related genes such as
often in intragenic regions and distal to classic CpG aromatase. In contrast, it was found that silencing
islands, the methylation of which is typically negatively GATA significantly reduced the established markers
correlated with gene expression (Fig. ) (). A total of of decidualization (Fig. ) (). In the absence of
 genes showed significant differences in methyla- GATA, the induction of HAND and prolactin in
tion; a large proportion of these genes encode tran- response to an in vitro decidualization (IVD) cocktail
scription factors (Fig. ) (). Many of these genes are including a progestin was reduced approximately by
implicated in the pathology of endometriosis and half, and IGFBP induction was strikingly reduced by
decidualization. Differential methylation affected the % (Fig. ) (). These findings collectively suggest
HOX gene clusters, nuclear receptor genes, and the that GATA acts as a mediator and partner of pro-
GATA family of transcription factors (). It is now gesterone signaling in normal endometrial stromal
clear that DNA methylation differentially regulates the cells and may enhance the decidual response (Fig. )
expression of hundreds of genes in endometriotic and (, ).
endometrial stromal cells (Fig. ). Pathologic roles of GATA6 in endometri-
The eutopic endometrium of women with endo- osis. The ectopic expression of GATA in otherwise
metriosis also displays DNA methylation abnormalities normal eutopic endometrial stromal cells shifted gene
giving rise to altered gene expression and pro- expression to reflect GATA expression patterns seen
gesterone resistance (, ). In fact, this phe- in endometriosis (Fig. ) (). All three of the critical
nomenon was demonstrated in a mouse model of steroid receptors showed significant changes in ex-
endometriosis (, ). These findings support the pression (Fig. ). ERa mRNA was reduced by .-fold
hypothesis that, during the process of retrograde when GATA was overexpressed without IVD, and by
menstruation, epigenetically abnormal populations -fold after IVD (). The overexpression of GATA
of stromal cells in eutopic endometrium may be also reduced PR transcript levels an average of .-fold.

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1061


REVIEW

Figure 8. Genome-wide DNA methylation differences between stromal cells isolated from eutopic endometrium and endometriosis. The
methylated (C) or unmethylated (s) state of a cytosine nucleotide in the CpG (or CG) sequence may in part regulate how genes are
expressed in the DNA of a cell. (a) Schematic diagram of CpGs depicts their genomic context relative to the nearest CpG island (top) or
gene (bottom). Gene contexts of CpGs were described as within an “island” (brown); on the “shore,” defined as within 4 kb around the
island (yellow); or in “open sea,” defined as at least 4 kb distal from an island, and relative to the nearest open reading frame within 1500 bp
(TSS1500; purple) or 200 bp (TSS200; pink) of a transcription start site (TSS); in the 59 untranslated region (UTR), the first exon of a
transcript (green); in the body of the gene (orange) or the 39UTR (red). (b) Pie charts show the distribution of the CpGs examined based on
their genomic context for all probes retained from the original array (top), for all the probes identified as differentially methylated between
endometriosis and endometrium (middle), and for all the differentially methylated probes that were matched to differentially expressed

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
mRNAs (bottom; note that stacked bar graphs show the CpG island context broken down for each of the gene contexts). A large number
(5423) of differentially methylated CpG islands in this array were linked to a differentially expressed gene. A further integrative analysis
demonstrated that 403 genes associated with one or more differentially methylated CpGs are differentially expressed with possible
functional consequences. “Transcription factors” as a category comprised the most important gene category, whereas the top biologic
pathway was “blood vessel development.” [Reproduced from Dyson MT, Roqueiro D, Monsivais D, et al. Genome-wide DNA methylation
analysis predicts an epigenetic switch for GATA factor expression in endometriosis. PLoS Genet. 2014;10(3):e1004158. Copyright © 2014 by
Dyson et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use,
distribution, and reproduction in any medium, provided the original work is properly cited.]

1062 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

Figure 9. An epigenetic switch from GATA2 to GATA6 in endometriosis. In endometrium, the GATA2 gene is characterized by a lack of
DNA methylation (s). In contrast, endometriosis has hypermethylated CpGs throughout a CpG island and proximal sequences within
the GATA2 promoter and throughout the gene. Several regions of the GATA6 gene are completely methylated in endometrium, whereas
methylation was absent within three CpG islands and their shores within the body of the gene. As a result, GATA2 is expressed in healthy
cells and upregulates several genes involved in decidualization; GATA2 may also maintain the expression of aldehyde dehydrogenase 1
family, member A2 (ALDH1A2), which is a key enzyme in retinoid metabolism. In endometriotic cells, aberrant methylation permits
expression of GATA6. GATA6 regulates the expression of several genes involved in steroid metabolism, the nuclear steroid hormone
receptors, and other GATA family members. Ectopic expression of GATA6 drives a pattern of gene expression similar to that seen in
endometriotic tissues, essentially transforming healthy endometrium away from spontaneous decidualization and toward the disease

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
phenotype. [Reproduced from Dyson MT, Roqueiro D, Monsivais D, et al. Genome-wide DNA methylation analysis predicts an epigenetic
switch for GATA factor expression in endometriosis. PLoS Genet. 2014;10(3):e1004158. Copyright © 2014 by Dyson et al. This is an open
access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited.]

In contrast, the expression of ERb increased an Endometriotic stromal cells synthesize estradiol via
average of .-fold; IVD did not significantly affect the steroidogenic pathway, whereas normal endo-
these genes (). The effects of GATA on the metrial stromal cells do not produce steroid hormones
protein levels of these steroid receptors have not (). SF (also known as nuclear receptor subfamily ,
been reported. Matrix metallopeptidase  (MMP) group A, member , or NRA) is critical, but not
and aromatase mRNA levels were also strikingly sufficient, for activating the cascade that involves at
altered by GATA overexpression (Fig. ). MMP least five steroidogenic proteins or enzymes (Fig. )
was repressed by IVD, but overexpression of (). Silencing of GATA in endometriotic stromal
GATA further reduced its transcript levels by cells showed that GATA was necessary for catalyzing
-fold relative to untreated controls. Aromatase the conversion of progesterone to androstenedione by
was expressed at very low levels basally, but was -hydroxylase/,-lyase (CYPA) (Fig. ).
strikingly increased after GATA overexpression (Fig. Ectopic expression of GATA alone or with SF was
) (). Overexpression of GATA profoundly re- essential for converting pregnenolone to estrogen
stricted the ability of endometrial stromal cells to [b-hydroxysteroid dehydrogenase- (HSDB),
decidualize, effectively blocking all four genes expected CYPA, and aromatase (CYPA)] (). How-
to increase with IVD (FOXO, HAND, PRL, and ever, simultaneous ectopic expression of both GATA
IGFBP; Fig. ) (). and SF was required and sufficient to confer

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1063


REVIEW

induction of all five genes and their encoded proteins ligand) that was first described as a regulator of
that convert cholesterol to estradiol (Fig. ) (). adrenal and gonadal steroid production (). It was
Functionally, only simultaneous knockdown of GATA the first nuclear receptor whose function was de-
and SF blocked estradiol formation in endometriotic fined in detail in endometriosis (). The practical
stromal cells (). The presence of both transcription absence of the orphan nuclear receptor SF in
factors was required and sufficient to transform en- endometrial stromal cells and its ,-fold higher
dometrial stromal cells into endometriotic-like cells that presence in endometriotic stromal cells is, in part,
produced estradiol in large quantities (Fig. ) (). determined by a classic CpG island within its

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
Thus, expression of both GATA and SF in endo- promoter. This CpG island is heavily methylated in
metrial stromal cells is sufficient for transformation into endometrial stromal cells, which allows recruitment
endometriotic-like cells capable of converting choles- of the silencer-type transcription factor methyl-
terol to estradiol (Fig. ) (). CpG–binding domain protein  that interferes with
the binding of transcriptional activators to the SF
SF1: coordinator of estrogen formation promoter (). In endometriotic cells, the transcription
in endometriosis factor upstream stimulatory factor- (USF) is able
Epigenetic regulation of the SF1 gene. SF is to bind to the unmethylated SF promoter to ac-
an orphan nuclear receptor (without a known tivate it (). In vivo, USF levels are strikingly

Figure 10. Estradiol production in endometriosis: roles of SF1 and GATA6. The endometriotic stromal cell is capable of converting
cholesterol to estradiol via a number of enzymatic steps. Both transcription factors, SF1 and GATA6, are essential for the production of
estradiol and mediate the effects of PGE2 in stimulating these steroidogenic genes. SF1 is essential and sufficient for the expression of at
least five steroidogenic genes, whereas SF1 and GATA6 together are essential and sufficient for the expression of three of these genes.
The effects of SF1 or GATA6 on HSD17B1 in endometriosis are not known. The addition (ectopic expression) of both SF1 and GATA6 is
required for a normal endometrial stromal cell to make estradiol from cholesterol. CYP11A1, side-chain cleavage enzyme; HSD3B2, 3b-
hydroxysteroid dehydrogenase-2; HSD17B1, 17b-hydroxysteroid dehydrogenase type 1.

1064 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

higher in endometriosis compared with endometrium. The PGE receptor subtypes EP, EP, EP, and EP
Thus, differential SF expression in endometriosis vs are expressed at similar levels in both endometriotic and
endometrium is primarily controlled by an epigenetic endometrial stromal cells (). Activation of the EP
mechanism that permits binding of activator vs in- receptor increases the levels of intracellular cAMP,
hibitor complexes to its promoter (, ). which induces STAR and aromatase expression in
As detailed above, SF and GATA play a key role in endometriotic stromal cells (, , ). STAR and
the conversion of cholesterol to estradiol in endo- aromatase levels and activity are stimulated by PGE
metriotic stromal cells (Fig. ) (). Estrogen supports or a cAMP analog in endometriotic cells but not in

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
endometriotic cell survival, whereas prostaglandins and endometrial stromal cells (Fig. ) (, , ). The
cytokines lead to inflammation, chronic pelvic pain, and transcription factor SF, which is present in endome-
infertility (, ). A positive feedback loop links in- triosis and absent in endometrium, mediates the PGE/
flammation and estrogen production in endometriotic cAMP-dependent induction of STAR, aromatase, and
tissue, favoring overexpression of key steroidogenic other steroidogenic genes in endometriotic stromal cells
genes, notably aromatase, overexpression of COX, and (Fig. ) (). In PGE-treated endometriotic cells, SF
continuous local production of estradiol and prosta- assembles into an enhancer transcriptional complex on
glandin E (PGE) (Fig. ) (, –). the STAR and aromatase promoters to induce gene
Tissue sources of estrogen in endometriosis. expression ().
Inhibition of estrogen action by GnRH analogs, oral The absence of SF in endometrial cells underlies
contraceptives, progestins, and aromatase inhibitors the lack of responsiveness of steroidogenic genes to
significantly reduces pelvic disease and pain (Fig. ) PGE. Additionally, a number of transcriptional in-
(). Estrogen production starts with the entry of hibitors of STAR and aromatase provides a fail-safe
cytosolic cholesterol into the mitochondrion facili- system for silencing these steroidogenic genes in en-
tated by STAR. Six enzymes [side chain cleavage dometrial cells. These repressors are chicken oval-
enzyme (CYPA), HSDB, CYPA, aromatase bumin upstream transcription factor (COUP-TF),
(CYPA), and b-hydroxysteroid dehydrogenase- Wilms’ tumor- (WT), and CAAT/enhancer binding
(HSDB)] catalyze the conversion of cholesterol to protein-b (C/EBPb). Normal endometrial tissue ex-
biologically active estradiol (Fig. ) (). Inhibition presses much higher levels of WT and C/EBPb
of aromatase (CYPA) effectively eliminates all compared with endometriotic tissue. In the absence of
estrogen production (Fig. ) (). SF coordinately SF, these repressors form a transcriptional complex
binds to the promoters of the genes that encode on the steroidogenic promoters that suppresses ex-
STAR, CYPA, HSDB, CYPA, and aromatase pression in endometrial cells ().
(CYPA), making it an essential transcription factor In summary, upon PGE  induction, SF is
regulating steroidogenesis in the ovary and endome- recruited to the promoters of the essential steroido-
triosis (Fig. ) (). genic genes to drive local estradiol synthesis in
Estrogen is produced at three major body sites in endometriotic stromal cells (Fig. ). Aromatase in-
women with endometriosis. First, estradiol is secreted hibitors completely block estradiol synthesis, making it
by the ovary and reaches endometriotic tissue via the an attractive target for endometriosis therapeutics.
circulation. Follicular rupture at each ovulation Aromatase inhibitors have been shown to diminish or
causes extraordinarily large amounts of estradiol to eradicate treatment-refractory endometriotic implants
be released directly onto pelvic implants. Second, and associated pain ().
aromatase produced by peripheral adipose and skin
tissue converts circulating androstenedione to es- ERb: mediator of estrogen-induced inflammation
trone, which may reach endometriotic tissue via Epigenetic regulation of the ESR2 and ESR1
the circulation. Endometriotic tissue expresses the genes. In endometriotic stromal cells, ERb (ESR)
complete set of steroidogenic genes, including aro- levels are -fold higher and ERa (ESR) levels are
matase, allowing local conversion of cholesterol to -fold lower compared with normal endometrium
estradiol (). Local aromatase activity in endo- (Fig. ) (). Hypomethylation of a CpG island at
metriotic tissues may underlie the observed superior the promoter region of the ESR gene leads to high
effect of treatment with a GnRH analog plus an levels of expression in endometriotic stromal cells, and
aromatase inhibitor compared with GnRH analog hypermethylation silences the ESR gene in endo-
alone for providing long-lasting pain relief in women metrial stromal cells (Fig. ). In contrast, the ESR
with endometriosis (Fig. ) (). promoter is unmethylated in eutopic endometrium
PGE2 stimulates estradiol formation. PGE and heavily methylated in endometriosis (Fig. ) (),
stimulates the expression of all steroidogenic genes in leading to lower ERa levels in endometriotic vs en-
endometrial stromal cells, leading to local de novo dometrial stromal cells (). Consequently, the ab-
synthesis of estradiol (). PGE regulation of STAR normally high ERb/ERa ratio in endometriotic
and aromatase has been characterized extensively (Fig. stromal cells may perturb estradiol induction of the
) (). PGR (PR) gene, which in turn may in part be the basis

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1065


REVIEW

Figure 11. Estrogen and PRs in endometriosis. Compared with signals at RERG leads to endometriotic cell proliferation.
endometrial stromal cells, ERb levels are higher and ERa levels The ERb/PGE/RERG pathway represents a novel
are lower in endometriotic stromal cells because of altered DNA candidate for therapeutic intervention (Fig. ) ().
methylation. The PR gene is also differentially methylated
Estradiol/ERb also stimulates SGK expression and
between these two cells types. It is likely that the severely
elevated ratio of ERb/ERa is in part responsible for the observed enzyme activity, leading to increased human endometriotic
inhibition of ERa and PR expression in endometriotic stromal cell survival (Fig. ) (). In uterine microvascular en-
cells. dothelial cells, ERb mediates estradiol-stimulated COX
expression and PGE production ().

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
In a mouse model of endometriosis that expresses
high ERb levels, treatment with an ERb-selective es-
tradiol antagonist suppressed mouse ectopic lesion
growth (Fig. ) (). ERb interaction with apoptotic
machinery in the cytoplasm to inhibit TNF-induced
apoptosis may evade endogenous immune surveillance
and contribute to cell survival (Fig. ) (). ERb also
enhances cellular adhesion and proliferation properties
by interacting with components of the cytoplasmic
inflammasome to increase IL-b (Fig. ) ().
ERb and PGE2 connect estrogen and in-
flammation. Pelvic pain and infertility in endome-
triosis are directly related to endometriotic cell survival
and inflammation. Estrogen enhances the survival or
persistence of ectopic endometriotic tissue, whereas
prostaglandins and cytokines mediate pain,
for low PR expression in endometriosis (Fig. ). In inflammation, and infertility (, ). Inflammation
endometrial vs endometriotic stromal cells, the PGR and estrogen production in endometriosis are linked
gene is also differentially methylated, which may by a positive feedback cycle in which chronic over-
contribute to its suppressed expression in endo- expression of aromatase and COX support sustained
metriotic cells (Fig. ) (). production of estradiol and PGE in endometriotic
ERb is the key mediator of estrogen action in tissue (Fig. ) (, –). Estradiol/ERb stimulates
endometriotic stromal cells. Endometriotic stromal cells PGE formation, whereas PGE stimulates estradiol
show severely suppressed ERa and significantly higher synthesis (, ). Selective or nonselective COX in-
ERb levels compared with eutopic endometrial stromal hibitors that disrupt PGE synthesis effectively reduce
cells (Fig. ) (). A hypomethylated promoter region pelvic pain in endometriosis (). Likewise, aroma-
in the ESR gene results in remarkably higher ERb tase inhibitors that disrupt estradiol biosynthesis
mRNA and protein expression relative to the normal significantly reduce endometriosis-associated pain and
endometrium (Fig. ) (, ). The eutopic endo- cause regression of pelvic lesions (, ).
metrium of women with endometriosis also shows Taken together, ERb plays a central role in
higher ERb expression compared with the endome- the development and pathophysiology of endometri-
trium of healthy women, suggesting that abnormally osis. The overproduction of estradiol in endometriosis
high levels of ERb in the endometrium may predispose drives ERb signaling to support endometriotic tissue
women to develop endometriosis (, ). In survival and inflammation (). Additionally, ERb may
endometriotic stromal cells, ERb was found to tran- have estradiol-independent pathologic actions.
scriptionally repress ERa, indicating that elevated ERb
confers a unique estrogen response mechanism that PR: progesterone resistance
may contribute to disease progression (Fig. ) (). Role of progesterone in endometriosis.
ERb action in endometriosis and uterine Compared with the clearly pathologic effect of es-
tissue. Genome-wide comparative analysis of ERb trogen in endometriosis, the role of progesterone
binding and gene expression in human endometriosis has remained ambiguous for several reasons. First,
and endometrial tissues identified Ras-like, estrogen- although progesterone has a protective role in endo-
regulated, growth inhibitor (RERG) and serum and metrial cancer (characterized by epithelial proliferation),
glucocorticoid–regulated kinase (SGK) as key ERb it does not have the same effect in endometriosis, in
targets (Fig. ) (, ). Estradiol induces RERG mRNA which benign lesions are comprised of primarily stro-
and protein levels in human endometriotic stromal cells, mal cells with diminished apoptosis or differentiation
whereas PGE phosphorylates RERG and enhances its (). Paradoxically, progesterone induces a transient
nuclear translocation. RERG induces ribosome bio- proliferation of stromal cells in normal endometrium
genesis and the proliferation of primary endometriotic during the secretory phase (). Second, progestin-
cells (Fig. ); thus, the integration of ERb and PGE based treatments are at best variably effective at

1066 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

reducing endometriosis-related pain, especially in pa- RAR-g and peroxisome proliferator-activated receptor
tients who received other forms of treatment (, ). (PPAR)-b/d, to regulate target gene transcription (Fig.
Progestins likely reduce pain via suppressing or attenu- ) (). Because STRA is strongly induced by PR
ating ovulation (), although a direct effect on endo- signaling, PR-deficient endometriotic stromal cells are
metriotic tissue cannot be excluded. Third, antiprogestins unable to produce sufficient RA (Fig. ) (, ).
with mixed agonist and antagonist properties (also Transcriptional activation of the nuclear receptor
known as selective PR modulators) reduce RAR by RA often leads to cell growth inhibition.
endometriosis-associated pelvic pain, possibly more However, in some tissues, RA acting through PPARb/

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
effectively than progestins (, , ). Furthermore, d induces the expression of prosurvival genes to
endometriotic tissues produce significant quantities of promote cell survival and hyperplasia (). RA ac-
progesterone and express significantly lower levels of tivation of these two receptors is regulated by the
PR compared with endometrium (, ). These intracellular lipid-binding proteins CRABP and
conflicting observations have prevented the formation FABP, which specifically deliver RA from the cytosol
of a unified hypothesis describing progesterone’s role to nuclear RAR and PPARb/d, respectively ().
in endometriosis. Thus, RA signals through RAR to drive apoptosis in
PR deficiency in endometriosis. Progesterone cells with a high CRABP/FABP ratio, but signals
resistance in endometriotic tissue is readily explained through PPARb/d to promote survival in cells with a
by the extremely low PR levels (). In normal en- low CRABP/FABP ratio ().
dometrium, levels of the PR isoforms, PR-B and In normal endometrial stromal cells, RA induces
PR-A, increase during the proliferative phase, peak just apoptosis via the CRABP/RARa pathway; in
before ovulation, and then diminish; this pattern CRABP/RARa-deficient endometriotic cells, RA
suggests that estradiol stimulates PR expression (). does not have the same effect (Fig. ) (, ). This
In simultaneously collected tissues of endometriosis, favors survival of endometriotic stromal cells and may
PR-B is undetectable and PR-A is markedly lower support the development and persistence of pelvic
compared with normal endometrium. Moreover, the endometriotic implants. Because PR induces CRABP
endometrium of baboons with endometriosis shows gene expression (Fig. ) (), PR resistance in
deficiency of the co-chaperone FKBP, which is endometriotic stromal cells leads to deficiency of
necessary for PR function, and this may contribute to CRABP; this supports the overall model of deficient
progesterone resistance (). PR action in endometriosis (Fig. ) ().
17b-Hydroxysteroid dehydrogenase-2 en- General deficiency of progesterone action in
zyme deficiency in endometriosis. Progesterone endometriosis. Estrogen and progesterone are es-
has an antiestrogenic effect in the healthy endometrium, sential and sufficient for endometrial function by
but a hallmark of endometriosis is progesterone re- regulating expression of multiple genes during the
sistance and elevated local estradiol levels ().
b-Hydroxysteroid dehydrogenase type (HSDB)
catalyzes the conversion of biologically potent estradiol
to less potent estrone (Fig. ) (–). Progesterone
via PR increases formation of retinoic acid (RA) in
endometrial stromal cells, which induces HSDB
expression in neighboring endometrial epithelial cells
(Fig. ) (–). In contrast, endometriotic
stromal cells with progesterone resistance do not
produce RA (Fig. ) (), which leads to a loss of
paracrine signaling to induce HSDB expression in
the epithelial cells and therefore failure to inactivate
estradiol in endometriosis (Figs.  and ) (, ).
Combined with high estradiol production due to
aberrant aromatase activity, this additional defect
further contributes to the abnormally high estradiol
activity in endometriosis (Figs.  and ) ().
Retinoid deficiency in endometriosis. Reti-
nol binding protein binds retinol (vitamin A) and Figure 12. ERb action in endometriosis. ERb induces RERG expression, whereas PGE2, via protein
delivers it to tissues via the circulation (Fig. ) (, kinase A (PKA), phosphorylates RERG. RERG phosphorylation is associated with its nuclear
translocation. Estradiol- and PGE2-mediated activation of RERG regulates endometriotic cell
). Retinol binds its cell surface receptor STRA
proliferation. Estradiol via ERb also induces expression of serum and glucocorticoid–regulated
and enters the cell, where retinol and retinal de- kinase (SGK1), which is also activated by PGE2 and oxidative stress. This leads to phosphorylation of
hydrogenase enzymes catalyze its conversion to FOXO3a, a proapoptotic factor, and enhanced cell survival. [Adapted from Monsivais D, Dyson MT,
transcriptionally active RA. RA acts through various Yin P, et al. ER beta- and prostaglandin E2-regulated pathways integrate cell proliferation via Ras-like
RA receptors (RARs), including RAR-a, RAR-b, and and estrogen-regulated growth inhibitor in endometriosis. Mol Endocrinol. 2014;28(8):1304–1315.]

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1067


REVIEW

menstrual cycle (). Indeed, administration of es- progesterone action has tangible downstream
tradiol and progesterone is sufficient to prepare the consequences. Upon exposure to progesterone in
endometrium for implantation in postmenopausal endometrial capillaries, poorly differentiated stromal
women undergoing donor embryo transfer (). cells with deficient PR fail to send physiologic para-
Progesterone exposure induces differentiation of both crine signals to adjacent epithelial cells (). RA is an
endometrial stromal cells (decidualization) and epi- essential paracrine factor, and its deficiency due to
thelial cells (secretory phenotype). Increased epithelial progesterone resistance is associated with the de-
glycodelin and stromal prolactin levels signal pro- ficiency of the enzyme HDSB in endometrial

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
gesterone action in the endometrium (, , epithelial cells (, , ). HSDB is essential
). Progesterone induces lower levels of prolactin for effectively inactivating local estradiol at the en-
expression in endometriotic vs endometrial stromal dometrial epithelium–embryo interface during the
cells, again supporting the progesterone resistance implantation window (). HSDB deficiency be-
model in endometriosis (). cause of progesterone resistance gives rise to local
Gene expression profiles in the endometrium of estradiol excess in endometriotic tissue (). This
women with or without endometriosis revealed that a possibly causes enhances estradiol-driven inflamma-
number of progesterone target genes are dysregulated tion and also implantation failure in eutopic endo-
during the window of implantation, the time when metrium because progesterone resistance was also
progesterone levels in the endometrium reach their demonstrated in eutopic endometrial tissue of patients
peak (, ). As an example, the progesterone- with endometriosis (). Interestingly, compounds
responsive gene glycodelin is downregulated in the with mixed progesterone antagonist/agonist properties
endometrium of women with endometriosis com- more effectively reduce uterine bleeding and pelvic
pared with women without endometriosis (). Thus, pain associated with endometriosis compared with
the eutopic endometrium of women with endome- progestins (, , ). This may be due to their
triosis also shows evidence of progesterone resistance ability to bind and act via lower levels of PR in
(, ). endometriotic tissue and/or by affecting the endo-
To summarize, progesterone resistance in endo- metrium (, , ).
metriosis is associated with PR deficiency in poorly
differentiated stromal cells (Fig. ). Defective
Nuclear receptor coregulators in endometriosis
Steroid receptor coactivators (SRCs) dynamically mod-
ulate nuclear receptor–mediated cellular processes in a
tissue-selective manner to precisely respond to ex-
ternal hormonal stimuli (). Alterations in SRC
concentrations or modifications of SRCs in steroid
target tissues may underlie nuclear receptor–
dependent human disease progression (). An
SRC-null mouse model revealed that the SRC gene
plays an essential role in endometriosis progression
(). Compared with normal endometrium, a -kDa
proteolytic C-terminal isoform of SRC is highly el-
evated in both the endometriotic tissue of mice with
surgically induced endometriosis and in eutopic en-
dometrial stromal cells biopsied from patients with
endometriosis (). TNF-induced MMP activity
mediates formation of the -kDa SRC isoform in
endometriotic mouse tissue. In contrast to full-length
SRC, the endometriotic SRC fragment prevents
TNF-mediated apoptosis in human endometrial ep-
ithelial cells and causes the epithelial–mesenchymal
transition and the invasion of human endometrial
Figure 13. Interactions of ERb, TNF, SRC1, and IL-1b in endometriosis. ERb plays a unique role cells (). Thus, the TNF/MMP/SRC pathway
in endometriotic tissue, where it interacts with the cytoplasmic apoptotic machinery and promotes key pathologic functions in endometriotic
inflammasome complex to prevent TNF-induced cell death and enhance adhesion and proliferative tissue (Fig. ) ().
activities of endometriotic tissues via SRC1 and IL-1b in this broad mechanistic pathway. This
The nuclear actions of the SRC fragment are
nongenomic action of ERb has a predominant role in endometriosis progression. [Reproduced
from Han SJ, Jung SY, Wu SP, Hawkins SM, Park MJ, Kyo S, Qin J, Lydon JP, Tsai SY, Tsai MJ, DeMayo
currently not known (). It is possible that SRC
FJ, O’Malley BW. Estrogen receptor b modulates apoptosis complexes and the inflammasome to or other nuclear receptor coregulators modify the
drive the pathogenesis of endometriosis. Cell. 2015;163(4):960–974. doi: 10.1016/j.cell.2015.10.034. activities of key nuclear receptors, such as ERb, SF,
Copyright © 2015 by Elsevier, Inc.] and PR, in endometriotic stromal cells (Fig. ).

1068 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

Summary of key mechanisms in endometriosis ERb mediates estrogen action in endometriosis.


Women who develop the clinical symptoms of en- Estradiol acts via ERb to stimulate COX, leading to
dometriosis may have an inheritable predisposition to overproduction of PGE. Thus, inflammation and
this disease, including epigenomic abnormalities in estrogen are linked via a positive feedback cycle in-
tissue progenitor/stem cells in the coelomic epithelium volving overexpression of genes encoding the aro-
(peritoneum), endometrium, endometrial vasculature, matase and COX enzymes and continuous formation
and other pelvic tissues. Continual and frequent ep- of their products, estradiol and PGE, in endometriotic
isodes of ovulatory menses that expose the lower tissue. Moreover, a strikingly low ERa/ERb ratio is

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
abdominal cavity to large quantities of blood and shed associated with deficient induction of PR levels, which
endometrial tissue increase the risk for developing leads to progesterone resistance and disruption of a
pelvic endometriosis. Exposure of pelvic tissues to paracrine pathway that inactivates estradiol. Enhanced
high levels of estradiol also increases the risk and estradiol formation and deficient inactivation in en-
severity of endometriosis, driving the persistence, dometriosis results in accumulation of estradiol. Fi-
tissue remodeling, and inflammation in endometriotic nally, via genomic and nongenomic pathways, ERb
lesions (Fig. ). modulates the activities of RERG, TNF, and IL-b to
Repeated exposure of lower abdominal tissues to promote survival, proliferation, and inflammation in
poorly differentiated endometrial mesenchymal stem poorly differentiated endometrial stromal cells. This
cells gives rise to the formation of symptomatic en- model is clinically relevant because therapeutic tar-
dometriosis in the peritoneum, ovarian surface and geting of aromatase, COX, ERb, or PR reduces pelvic
hemorrhagic cysts, and the area between the rectum pain or ablates visible endometriotic lesions (Figs. ,
and vagina. The endometriotic stromal cell displays , and ).
critical abnormalities in genome-wide DNA methyl- It is now clear that the epithelial cells of normally
ation, regulation of gene expression, and signaling functioning endometrium contain numerous
pathways. Most of these perturbations occur in
stromal cells exhibiting stem cell properties and partial
ovarian granulosa cell–like and immune cell–like
characteristics such as estradiol, prostaglandin, and
cytokine production. These cells originate from the
eutopic endometrium and exhibit abnormal expres-
sion of key transcription factors, including high levels
of GATA, SF, and ERb and deficiencies in GATA,
ERa, and PR in both eutopic and ectopic stromal cells
(Figs.  and ).
GATA and ERa regulate key genes necessary
for progesterone-driven differentiation of healthy
endometriotic stromal cells. The promoters of these
genes are hypermethylated and thus transcriptionally
repressed in endometriotic cells, whereas GATA is
hypomethylated and abundant in endometriotic
cells. GATA blocks hormone sensitivity, represses
GATA, and induces markers of endometriosis when
expressed in healthy endometrial cells. Ectopic ex-
pression of GATA in a normal endometrial stromal
cell effectively transforms it to an endometriotic
stromal cell, recapitulates key molecular defects such
as aromatase and ERb expression, and induces es-
trogen biosynthesis and progesterone resistance
(Figs.  and ).
The sustained cell survival and inflammation
characteristic of endometriosis have been linked to Figure 14. Progesterone resistance: HSD17B2 deficiency in endometriotic epithelium. HSD17B2 is
a stromal cell defect involving excessive formation specifically expressed in eutopic endometrial epithelial cells during the secretory phase. This
of estrogen and prostaglandin and progesterone re- expression coincides with progesterone secretion by the corpus luteum. HSD17B2 converts the
sistance, all of which originate from two transcription biologically potent estrogen, estradiol (E2), to estrogenically weak estrone (E1). In endometriotic
tissue, however, this epithelial HSD17B2 is severely deficient, giving rise to E2 excess. These findings
factors, SF and ERb. Exposure of endometriotic cells
are suggestive that progesterone stimulates HSD17B2 in eutopic endometrium and that its
to the local hormone PGE leads to coordinated deficiency in endometriosis is a consequence of progesterone resistance. [Reproduced from
binding of SF to the promoters of multiple ste- Zeitoun K, Takayama K, Sasano H, Suzuki T, Moghrabi N, Andersson S, Johns A, Meng L, Putman M,
roidogenic genes, including aromatase, causing for- Carr B, Bulun SE. Deficient 17b-hydroxysteroid dehydrogenase type 2 expression in endometriosis:
mation of large quantities of estradiol (Fig. ). failure to metabolize 17b-estradiol. J Clin Endocrinol Metab. 1998;83(12):4474–4480.]

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1069


REVIEW

Figure 15. Paracrine stromal–epithelial interactions for progesterone and retinoid action in endometrium and endometriosis.
Deficient genes and pathways in endometriosis are indicated by arrows and dotted lines. Blood vessels deliver progesterone (P4) to
endometrial stromal cells, which express PR; activation leads to the production of paracrine factors, including RA. RA acts in a paracrine
manner to stimulate differentiation and oppose estradiol (E2)-dependent proliferation in endometrial epithelial cells. Moreover, RA
stimulates HSD17B2, which converts biologically active E2 to estrogenically weak estrone (E1). Endometriotic stromal cells express
lower levels of PR, which leads to lower RA formation. As a result, paracrine signaling to neighboring epithelial cells is lost, and these cells
do not differentiate or express HSD17B2, leading to excess E2. The mechanisms supporting retinoid transport between endometrial
stromal and epithelial cells are not fully understood. The retinoid cell surface receptor stimulated by RA-6 (STRA6) in endometrial
stromal cells binds retinol binding protein (RBP)–bound retinol in the circulation for cellular uptake of retinol. Retinol is converted to

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
RA and transported to nuclear RARs by the shuttling protein cellular RA binding protein-2 (CRABP2). RA-bound RAR stimulates
endometrial stromal cell differentiation and apoptosis. PR induces stromal STRA6 and CRABP2 expression. In endometriosis, this
pathway is disrupted by a deficiency of PR, STRA6, and CRABP2 as abnormal expression of the RA-metabolizing enzymes CYP26B1 and
CYP26A1 in stromal and epithelial endometrial cells. 4OH-RA, 4-hydroxy-RA. [Reproduced from Kim JJ, Kurita T, Bulun SE. Progesterone
action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocrine Rev. 2013;34(1):130–162.]

mutations, including driver mutations of ovarian misprogrammed endometriotic cells, may enhance
cancer. Some of these mutations (e.g., in KRAS, epithelial mutagenesis or carcinogenesis [Fig. 
PIKCA, and ARIDA) are enriched in endometriotic ()].
epithelial cells from ovarian endometriomas or Some intriguing questions remain unanswered.
extraovarian deep-infiltrating endometriosis and What are the relative roles of the environment vs the
may play roles in the pelvic implantation of endo- genome in the widespread epigenomic abnormalities
metriotic tissue fragments with survival and growth in endometriotic stromal cells? Are these epigenomic
advantages. Although epithelial driver mutations abnormalities transmissible through the generations?
in extraovarian sites do not cause malignant What happens to the epithelial cells carrying cancer
transformation, the same mutations in ovarian driver mutations in eutopic endometrium? What
endometriomas may be essential for initiating determines the malignant transformation of some but
endometriosis-associated epithelial ovarian can- not all ovarian endometriomas with similar epithelial
cers. The intense paracrine inflammatory sig- mutations? How do epigenomically abnormal stro-
nals, together with estrogen from epigenomically mal cells and mutated epithelial cells influence each

1070 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

other in a paracrine fashion in benign and malignant of elagolix for the management of endometriosis-
settings? associated pain have been demonstrated in phase 
and phase  trials (). During a -week period,
elagolix demonstrated similar efficacy on endometriosis-
Innovations in Clinical Management associated pain to subcutaneous medroxyprogesterone
acetate but with a minimal effect on bone mineral
The goals of medical therapy for endometriosis are pain density ().
control, improvement of the quality of life, prevention Phase  trials showed that higher ( mg twice a

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
of disease recurrence, fertility preservation, and re- day) and lower ( mg daily) doses of elagolix
duction of operative intervention (). Endometriosis- were effective in improving dysmenorrhea and
associated pain involves the stimulation of sensory nonmenstrual pelvic pain during a -month period in
nerve tissue in the eutopic endometrium, pelvic peri- women with endometriosis-associated pain ().
toneum, and other pelvic tissues by inflammatory Both doses of elagolix were, however, associated
substances produced as a consequence of the complex with hypoestrogenic adverse effects (). Extension
disease process. Inflammation is closely linked to re- studies evaluated  continuous months of treatment
peated episodes of ovulatory menses and retrograde with both doses of elagolix (). Endometriosis
travel of blood and other menstrual tissue into the pelvic patients benefited from sustained reductions in
cavity. Estrogen is the most hierarchically upstream and dysmenorrhea, nonmenstrual pelvic pain, and dys-
potent stimulus of survival and inflammation in eutopic pareunia (). The higher dose was associated with
and ectopic endometrial tissues. greater improvement of dysmenorrhea (). The
Following from this mechanistic understanding side effects were consistent with reduced estrogen
of disease pathology, successful treatments of levels and no new concerns were associated with
endometriosis-associated pain focus on suppression of long-term use of elagolix (). Nonhormonal
ovulation and estrogen production (). The ratio- contraception is needed, as neither dose of elagolix
nale for hormone therapy is to induce amenorrhea, reliably suppresses ovulation ().
thereby creating a relatively hypoestrogenic envi-
ronment that will inhibit the inflammatory process Aromatase inhibitors
and prevent disease progression (). Traditionally, Most hormonal treatments for endometriosis are
ovulatory cycles associated with estrogen secretion and focused on inhibiting ovarian estrogen production
retrograde menstruation have been suppressed at the rather than blocking estrogen produced locally in
level of hypothalamus/pituitary using oral contra- endometriotic lesions. Aromatase is encoded by a
ceptives, progestins, or GnRH agonists (). Pain single gene and is the final enzyme in the estrogen
and other abdominal symptoms usually recur upon biosynthesis pathway. Its inhibition effectively
treatment discontinuation, however (Fig. ), and these eliminates all estrogen production. Recently in-
treatments do not address pathogenic factors in en- troduced highly specific aromatase inhibitors have
dometriosis tissue such as local aromatase activity, successfully treated pelvic pain and significantly re-
nuclear receptor activity, or cytokine production. duced lesion size (). In premenopausal women, an
Some recently approved, experimental, or emerging aromatase inhibitor alone may induce ovarian fol-
treatments are discussed below. liculogenesis, and thus aromatase inhibitors are
combined with a progestin, a combination oral
Oral GnRH antagonists contraceptive, or a GnRH agonist (Fig. ) ().
Although GnRH agonists have been used to suppress Although combining an aromatase inhibitor with a
ovulation in the treatment of endometriosis, GnRH GnRH agonist may lead to extremely severe hypo-
antagonists have not been available until recently. Oral estrogenic side effects, the side-effect profile of aro-
nonpeptide forms of GnRH antagonists (e.g., elagolix, matase inhibitors administered in combination with
relugolix) have been developed, and elagolix has been an oral contraceptive or a progestin is remarkably
approved by the Food and Drug Administration to tolerable and consists of mild hot flashes and de-
treat endometriosis (, ). Elagolix is an oral creased sexual desire (). In premenopausal pa-
GnRH antagonist that rapidly suppresses the pituitary- tients, the combination of an oral contraceptive or
ovarian hormones in a dose-dependent manner (). progestin (norethindrone acetate, . to  mg/d) with
However, ovulation may still occur during treatment, an aromatase inhibitor reduces visible lesions and
particularly at lower doses that partially suppress pelvic pain refractory to other available medical and
ovarian estrogen production (). Oral GnRH an- conservative surgical treatments (–, ).
tagonists produce a hypoestrogenic environment; Endometriosis that persists after surgically in-
however, at low doses they can maintain sufficient duced or natural menopause is rather uncommon
circulating estradiol levels to avoid severe vasomotor but continues to be a serious clinical challenge ().
symptoms, vaginal atrophy, and bone demineraliza- For postmenopausal endometriosis, the medical
tion (, ). The efficacy, safety, and tolerability treatment of choice is an aromatase inhibitor, either

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1071


REVIEW

anastrozole ( mg/d) or letrozole (. mg/d) (). Hot association with its use (). UPA was not taken off the
flashes and bone loss may be mitigated by adding market, but the European Medicines Agency has ad-
norethindrone acetate (. to  mg/d) and/or a vised periodic liver monitoring before, during, and after
bisphosphonate (zoledronic acid,  mg IV infusion/yr). treatment with UPA in all patients who consider using it
Long-term use of a progestin may increase breast cancer for uterine fibroids ().
risk in a postmenopausal woman (). In contrast, in
postmenopausal women with breast cancer, zoledronic Targeting prostaglandin production or action
acid decreases the recurrence of invasive breast cancer Prostaglandins are locally produced hormones linked

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
but slightly increases the risk of jaw necrosis associated to inflammation and pain in endometriosis. PGE and
with a dental procedure (). In advanced stage prostaglandin F (PGF)a are produced excessively in
(American Society for Reproductive Medicine stage III uterine and endometriotic tissues (). PGFa in-
to IV) endometriosis, the combination of anastrozole duces vasoconstriction and uterine contractions that
and a GnRH agonist during postsurgical therapy de- may contribute to dysmenorrhea, whereas PGE may
creased pain recurrence in approximately half of pa- induce pain directly (). Reduction of prostaglandin
tients at the -year follow-up (). formation by nonselective COX inhibitors has
been shown to significantly decrease endometriosis
Antiprogestins associated pelvic pain (, ). Chronic adminis-
Selective PR modulators have primarily anti- tration of nonselective COX inhibitors (e.g., ibuprofen,
progestogenic effects and include mifepristone (RU), naproxen) is limited due to their gastrointestinal side
asoprisnil, and UPA (). Mifepristone alleviates pain effects. Long-term administration of a selective COX
symptoms and induces amenorrhea without causing inhibitor with milder side effects significantly reduces
hypoestrogenism in patients with endometriosis (, chronic pelvic pain, but its use has been limited by
). UPA ( to  mg) is approved for the treatment of increased cardiovascular risk (). In fact, the COX-
endometriosis in Europe and Canada (). In clinical selective nonsteroidal anti-inflammatory drug celexocib
trials, anovulation was observed in .% women in the was approved by the Food and Drug Administration for
 mg group and % in the  mg group, and primary dysmenorrhea that is part of the symptom
amenorrhea occurred in .% and % of women in complex of endometriosis ().
the  mg and  mg groups, respectively (). Because Arachidonic acid released by phospholipase A is
UPA induces amenorrhea and possibly reduces pain, it presented to COX, which catalyzes its conversion to
can be used in cases of endometriosis with pain re- prostaglandin H (PGH) in most cells, including in
fractory to existing treatments (). After this drug was myometrium, endometrium, and endometriotic tissue
approved in Europe and Canada, sporadic cases of liver (). Among the two isoforms, COX is generally
injury and hepatic failure were recently reported in responsible for basal prostaglandin synthesis, whereas
COX is important in various inflammatory and in-
duced settings. The coupling of COX-dependent syn-
thesis of PGH to its metabolism by downstream
enzymes is orchestrated in a cell-specific fashion.
Uterine cells are rich in PGF synthase and microsomal
PGE synthase (mPGES), which catalyze the conversion
of PGH to PGFa and PGE, respectively ().
The contribution of excessive PGE production via
induction of multiple enzymes, in particular COX
and mPGES, in inflammatory settings such as endo-
metriosis is a developing concept (). Large quantities
of PGE produced by endometriotic stromal cells in turn
induce local estrogen biosynthesis and pelvic pain (, ,
, ). COX is upregulated in endometriotic stromal
cells compared with endometrial stromal cells, and its
Figure 16. Summary of key estrogen-dependent mechanisms in endometriosis. Pathologically
decreased methylation of the SF1, GATA6, and ERb genes causes extremely high levels of their proteins expression is also higher in endometrium of women with
in endometriotic stromal cells. SF1 together with GATA6 coordinately mediate the PGE2-induced endometriosis compared with endometrium of disease-
expression of many genes in the estrogen synthetic cascade to produce estradiol from cholesterol. In free women (, ). Expression of mPGES has been
addition to decreased methylation of its gene promoter, ERb expression is high also due to a observed in both benign and malignant endometrium
stimulatory effect from GATA6. Estradiol and ERb induce COX2 expression and PGE2 production in (–). Thus, increased synthesis of PGE observed in
endometrial cells. Moreover, PGE2 per se and IL-1b also induce COX2 expression and PGE2 production
vivo in endometriosis may be due to coordinate hy-
in endometrial cells. ERb and GATA6 suppress ERa and PR, leading deficiencies of RA production and
the HSD17B2 enzyme. The end result is extremely high local concentrations of estradiol and PGE2 in
peractivity of COX and mPGES.
endometriotic tissue; both of these molecules are key inducers of inflammation and pain in COX expression and PGE  production are
endometriosis. This pathway is clinically relevant because its disruption by an aromatase inhibitor or a stimulated in endometriotic and uterine cells (Fig. ).
COX inhibitor (e.g., a nonsteroidal anti-inflammatory drug) reduces the extent of disease or pelvic pain. The cytokine IL-b and PGE itself (in an autocrine

1072 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

manner) induce COX expression in endometriotic to ensure large quantities of PGE production. Estradiol
and endometrial stromal cells (, ). The angio- induces COX via ERb in endometriosis. Disruption of
genic factor VEGF and estradiol via ERb rapidly in- PGE synthesis via selective or nonselective COX in-
duce COX expression in uterine endothelial and hibitors effectively reduces pelvic pain in endometriosis
endometriotic stromal cells (–). These re- ().
dundant mechanisms maintain large quantities of
PGE in endometriotic tissue (Fig. ). Cytokines and ERb
The biological actions of PGE are mediated via the Estrogen and progesterone have variable effects on

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
G protein receptors EP, EP, EP, and EP, which the production of different cytokines in endometrial
integrate multiple cell signaling pathways (). or endometriotic cells in vitro and in vivo (, ).
Using a xenograft mouse model of endometriosis that Recently, an intriguing link was noted between ERb,
utilizes human endometriotic cell lines, pharmacological TNF, and IL-b in endometriotic tissue (). ERb
inhibition of the EP and EP receptors demonstrated acts as a master regulator that interacts with the
a potential clinical benefit (). Specifically, selective cytoplasmic apoptotic machinery and inflammasome
inhibition of EP/EP: (i) decreased growth and survival complex to prevent TNF-induced cell death and IL-
of endometriosis lesions; (ii) decreased angiogenesis b–enhanced adhesion and proliferative activities in
and innervation of endometriosis lesions; (iii) sup- mouse and human endometriotic tissues (). This
pressed the proinflammatory state of dorsal root ganglia nongenomic action of ERb has a predominant role in
neurons to decrease pelvic pain; (iv) blunted the pro- endometriosis progression (Figs.  and ), and
inflammatory, estrogen-dominant, and progesterone- ERb-selective compounds that act as estradiol an-
resistant molecular environment of the endometrium tagonists in endometriotic tissue are potential ther-
and endometriotic lesions; and (v) restored endometrial apeutics. The potential role for such a compound was
functional receptivity (). These data suggest that long- recently demonstrated in a mouse model of endo-
term inhibition of PGE action may be an alternative metriosis (). Likewise, the blockage of TNF action
treatment of endometriosis (). using systemically administered recombinant TNF
In summary, cytokines, angiogenic factors, PGE, and receptor type- or a monoclonal antibody against
estradiol all induce COX expression in endometriosis TNF prevented the establishment of endometriosis

Figure 17. A vision of the future of endometriosis treatment. Human iPS cells are currently being tested for the treatment of a number
of human conditions. The genomes of the biopsied skin or bone marrow cells are reprogrammed via the addition of four stem cell
factors (OCT4, SOX2, KLF4, and MYC) to generate iPS cells. It was recently demonstrated that these pluripotent stem cells can be
differentiated to endometrial stromal cells (ESCs) under defined molecular conditions (207). In the future, the differentiation protocol
may be optimized to produce appropriately progesterone-responsive and healthy ESCs, which can be used to replace epigenetically
defective and progesterone-resistant stromal cells in the intrauterine endometrial tissue of a woman with endometriosis.

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1073


REVIEW

or reduced the lesion size in a baboon model of Future treatment considerations


endometriosis (, ). Theoretically, replacement of abnormal eutopic en-
dometrial stromal cells with normal ones is a potential
Management of endometriosis-related infertility novel therapeutic approach for endometriosis. As
Infertility management in women with endometriosis is detailed in previous sections, epigenetically defective
challenging and controversial. Clinicians and patients and progesterone-resistant eutopic endometrial stro-
have moved away from laparoscopic surgical treatment mal cells with stem-like properties may reach the
of all stages of endometriosis, and many clinicians no pelvic cavity via retrograde menstruation and con-

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
longer operate on ovarian endometriomas until fertility tribute to the establishment and persistence of en-
treatments are completed (). In women with min- dometriosis (, , ). During the past decade, induced
imal to mild endometriosis (American Society for pluripotent stem (iPS) cells derived from bone marrow
Reproductive Medicine stage I to II), all of the fertility or skin biopsies provided a patient-specific source that
outcomes were comparable to disease-free women; this can be differentiated to various cells types and used
included live birth rate (). Women with more severe as a cell-based treatment (). It was also suggested
disease (American Society of Reproductive Medicine that normal endometrial stromal cells themselves
stage III to IV), however, had a lower live birth rate, could serve as a source of adult stem cells for thera-
lower clinical pregnancy rate, and lower mean number peutic applications ().
of oocytes retrieved per in vitro fertilization (IVF) cycle, A recent breakthrough opened up the possibility of
when compared with women with no endometriosis iPS cell–based treatment of endometriosis (Fig. ). A
(). Ovulation induction combined with intrauterine group directed the differentiation of human iPS cells
insemination seems to robustly increase the risk of through intermediate mesoderm, coelomic epithelium
recurrent endometriosis-associated pain or disease followed by the Müllerian duct to endometrial stromal
progression (). Therefore, IVF is the preferred fibroblasts under molecularly defined embryoid body
method for treating infertility associated with endo- culture conditions using specific hormonal treatments
metriosis (). In general, live birth rates associated (). Activation of b-catenin was essential for expression
with IVF are not different in infertility patients with or of PR that mediated the final differentiation step of
without endometriosis (), and pregnancy rates are endometrial stromal cells (). Thus, it is possible to use
not seemingly reduced in oocyte donation recipients skin fibroblasts of an endometriosis patient to generate
affected by endometriosis (). However, ovarian iPS cells and then epigenetically reprogram these iPS cells
suppression with a GnRH agonist or oral con- under defined hormonal and molecular conditions to
traceptive significantly improved implantation rates produce healthy and progesterone-responsive en-
in patients with endometriosis (). Ovarian sup- dometrial stromal cells. These healthy iPS-derived
pression before IVF should be considered in patients cells can be used for endometrial regeneration for
with advanced-stage endometriosis or adenomyosis. future cell-based treatments. Multiple major technical
Importantly, note that the clinical impact of endo- challenges need to be overcome before it may be
metrial factor infertility in endometriosis is still possible to replace diseased endometrial stromal cells
debated and not well understood. with the normal ones regenerated from iPS cells.

References and Notes


1. Bulun SE. Endometriosis. N Engl J Med. 2009;360(3): 6. Sampson JA. Peritoneal endometriosis due to the 12. Brosens I, Gordts S, Benagiano G. Endometriosis in
268–279. menstrual dissemination of endometrial tissue into adolescents is a hidden, progressive and severe
2. Vercellini P, Viganò P, Somigliana E, Fedele L. En- the peritoneal cavity. Am J Obstet Gynecol. 1927; disease that deserves attention, not just compas-
dometriosis: pathogenesis and treatment. Nat Rev 14(4):422–469. sion. Hum Reprod. 2013;28(8):2026–2031.
Endocrinol. 2014;10(5):261–275. 7. Giudice LC. Endometriosis. N Engl J Med. 2010; 13. Aredo JV, Heyrana KJ, Karp BI, Shah JP, Stratton P.
3. Noble LS, Simpson ER, Johns A, Bulun SE. Aromatase 362(25):2389–2398. Relating chronic pelvic pain and endometriosis to
expression in endometriosis. J Clin Endocrinol Metab. 8. Gargett CE, Schwab KE, Deane JA. Endometrial signs of sensitization and myofascial pain and
1996;81(1):174–179. stem/progenitor cells: the first 10 years. Hum Reprod dysfunction. Semin Reprod Med. 2017;35(1):88–97.
4. Noble LS, Takayama K, Zeitoun KM, Putman JM, Update. 2016;22(2):137–163. 14. Vercellini P, Somigliana E, Vigano P, Abbiati A, Barbara
Johns DA, Hinshelwood MM, Agarwal VR, Zhao Y, 9. Brawn J, Morotti M, Zondervan KT, Becker CM, G, Fedele L. “Blood on the Tracks” from corpora lutea
Carr BR, Bulun SE. Prostaglandin E2 stimulates Vincent K. Central changes associated with chronic to endometriomas. BJOG. 2009;116(3):366–371.
aromatase expression in endometriosis-derived pelvic pain and endometriosis. Hum Reprod Update. 15. Khan KN, Fujishita A, Kitajima M, Hiraki K,
stromal cells. J Clin Endocrinol Metab. 1997;82(2): 2014;20(5):737–747. Nakashima M, Masuzaki H. Occult microscopic
600–606. 10. Davis AC, Goldberg JM. Extrapelvic endometriosis. endometriosis: undetectable by laparoscopy in
5. Han SJ, Jung SY, Wu SP, Hawkins SM, Park MJ, Kyo S, Semin Reprod Med. 2017;35(1):98–101. normal peritoneum. Hum Reprod. 2014;29(3):
Qin J, Lydon JP, Tsai SY, Tsai MJ, DeMayo FJ, 11. Chen LC, Hsu JW, Huang KL, Bai YM, Su TP, Li CT, 462–472.
O’Malley BW. Estrogen receptor b modulates ap- Yang AC, Chang WH, Chen TJ, Tsai SJ, Chen MH. 16. Matsuzaki S, Canis M, Pouly JL, Wattiez A, Okamura
optosis complexes and the inflammasome to drive Risk of developing major depression and anxiety dis- K, Mage G. Cyclooxygenase-2 expression in deep
the pathogenesis of endometriosis. Cell. 2015; orders among women with endometriosis: a longitu- endometriosis and matched eutopic endometrium.
163(4):960–974. dinal follow-up study. J Affect Disord. 2016;190:282–285. Fertil Steril. 2004;82(5):1309–1315.

1074 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

17. Soysal S, Soysal ME, Ozer S, Gul N, Gezgin T. The expressed and -secreted mRNA expression in normal 47. Li X, Zhang Y, Zhao L, Wang L, Wu Z, Mei Q, Nie J, Li
effects of post-surgical administration of goserelin endometrium and endometriotic implants: assessment X, Li Y, Fu X, Wang X, Meng Y, Han W. Whole-
plus anastrozole compared to goserelin alone in of autocrine/paracrine regulation by in situ hybridization. exome sequencing of endometriosis identifies fre-
patients with severe endometriosis: a prospective Am J Pathol. 2001;158(6):1949–1954. quent alterations in genes involved in cell adhesion
randomized trial. Hum Reprod. 2004;19(1):160–167. 32. Bulun SE, Monsavais D, Pavone ME, Dyson M, Xue and chromatin-remodeling complexes. Hum Mol
18. Ailawadi RK, Jobanputra S, Kataria M, Gurates B, Q, Attar E, Tokunaga H, Su EJ. Role of estrogen Genet. 2014;23(22):6008–6021.
Bulun SE. Treatment of endometriosis and chronic receptor-b in endometriosis. Semin Reprod Med. 48. Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K,
pelvic pain with letrozole and norethindrone ac- 2012;30(1):39–45. Zeng T, Senz J, McConechy MK, Anglesio MS, Kalloger
etate: a pilot study. Fertil Steril. 2004;81(2):290–296. 33. Monsivais D, Dyson MT, Yin P, Navarro A, Coon JS, SE, Yang W, Heravi-Moussavi A, Giuliany R, Chow C,
19. Amsterdam LL, Gentry W, Jobanputra S, Wolf M, Pavone ME, Bulun SE. Estrogen receptor b regulates Fee J, Zayed A, Prentice L, Melnyk N, Turashvili G,

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
Rubin SD, Bulun SE. Anastrazole and oral contra- endometriotic cell survival through serum and Delaney AD, Madore J, Yip S, McPherson AW, Ha G,
ceptives: a novel treatment for endometriosis. Fertil glucocorticoid-regulated kinase activation. Fertil Bell L, Fereday S, Tam A, Galletta L, Tonin PN,
Steril. 2005;84(2):300–304. Steril. 2016;105(5):1266–1273. Provencher D, Miller D, Jones SJ, Moore RA, Morin GB,
20. Abushahin F, Goldman KN, Barbieri E, Milad M, 34. Monsivais D, Dyson MT, Yin P, Coon JS, Navarro A, Oloumi A, Boyd N, Aparicio SA, Shih IeM, Mes-Masson
Rademaker A, Bulun SE. Aromatase inhibition for Feng G, Malpani SS, Ono M, Ercan CM, Wei JJ, AM, Bowtell DD, Hirst M, Gilks B, Marra MA,
refractory endometriosis-related chronic pelvic Pavone ME, Su E, Bulun SE. ERb- and prostaglandin Huntsman DG. ARID1A mutations in endometriosis-
pain. Fertil Steril. 2011;96(4):939–942. E2-regulated pathways integrate cell proliferation associated ovarian carcinomas. N Engl J Med. 2010;
21. Vercellini P, Eskenazi B, Consonni D, Somigliana E, via Ras-like and estrogen-regulated growth inhibitor 363(16):1532–1543.
Parazzini F, Abbiati A, Fedele L. Oral contraceptives in endometriosis. Mol Endocrinol. 2014;28(8): 49. Kuo KT, Mao TL, Jones S, Veras E, Ayhan A, Wang
and risk of endometriosis: a systematic review and 1304–1315. TL, Glas R, Slamon D, Velculescu VE, Kuman RJ, Shih
meta-analysis. Hum Reprod Update. 2011;17(2): 35. Bruner KL, Matrisian LM, Rodgers WH, Gorstein F, IeM. Frequent activating mutations of PIK3CA in
159–170. Osteen KG. Suppression of matrix metal- ovarian clear cell carcinoma. Am J Pathol. 2009;
22. Heidemann LN, Hartwell D, Heidemann CH, loproteinases inhibits establishment of ectopic le- 174(5):1597–1601.
Jochumsen KM. The relation between endome- sions by human endometrium in nude mice. J Clin 50. Suvitie PA, Hallamaa MK, Matomäki JM, Mäkinen JI,
triosis and ovarian cancer—a review. Acta Obstet Invest. 1997;99(12):2851–2857. Perheentupa AH. Prevalence of Pain Symptoms
Gynecol Scand. 2014;93(1):20–31. 36. Langoi D, Pavone ME, Gurates B, Chai D, Fazleabas Suggestive of Endometriosis Among Finnish Ado-
23. Wilbur MA, Shih IM, Segars JH, Fader AN. Cancer A, Bulun SE. Aromatase inhibitor treatment limits lescent Girls (TEENMAPS Study). J Pediatr Adolesc
implications for patients with endometriosis. Semin progression of peritoneal endometriosis in baboons. Gynecol. 2016;29(2):97–103.
Reprod Med. 2017;35(1):110–116. 51. Rogers PA, D’Hooghe TM, Fazleabas A, Gargett CE,
Fertil Steril. 2013;99(3):656–662.e3.
24. Anglesio MS, Papadopoulos N, Ayhan A, Nazeran Giudice LC, Montgomery GW, Rombauts L,
37. Fang Z, Yang S, Gurates B, Tamura M, Simpson E,
TM, Noë M, Horlings HM, Lum A, Jones S, Senz J, Salamonsen LA, Zondervan KT. Priorities for en-
Evans D, Bulun SE. Genetic or enzymatic disruption of
Seckin T, Ho J, Wu RC, Lac V, Ogawa H, Tessier- dometriosis research: recommendations from an
aromatase inhibits the growth of ectopic uterine
Cloutier B, Alhassan R, Wang A, Wang Y, Cohen JD, international consensus workshop. Reprod Sci. 2009;
tissue. J Clin Endocrinol Metab. 2002;87(7):3460–3466.
Wong F, Hasanovic A, Orr N, Zhang M, Popoli M, 16(4):335–346.
38. McLaren J, Prentice A, Charnock-Jones DS, Millican
McMahon W, Wood LD, Mattox A, Allaire C, Segars 52. Schliep KC, Mumford SL, Peterson CM, Chen Z,
SA, Müller KH, Sharkey AM, Smith SK. Vascular
J, Williams C, Tomasetti C, Boyd N, Kinzler KW, Gilks Johnstone EB, Sharp HT, Stanford JB, Hammoud AO,
endothelial growth factor is produced by peritoneal
CB, Diaz L, Wang TL, Vogelstein B, Yong PJ, Sun L, Buck Louis GM. Pain typology and incident
fluid macrophages in endometriosis and is regu-
Huntsman DG, Shih IM. Cancer-associated endometriosis. Hum Reprod. 2015;30(10):2427–2438.
lated by ovarian steroids. J Clin Invest. 1996;98(2):
mutations in endometriosis without cancer. N Engl J 53. Apostolopoulos NV, Alexandraki KI, Gorry A, Coker
482–489.
Med. 2017;376(19):1835–1848. A. Association between chronic pelvic pain
39. Takayama K, Zeitoun K, Gunby RT, Sasano H, Carr
25. Suda K, Nakaoka H, Yoshihara K, Ishiguro T, Tamura symptoms and the presence of endometriosis. Arch
BR, Bulun SE. Treatment of severe postmenopausal
R, Mori Y, Yamawaki K, Adachi S, Takahashi T, Kase Gynecol Obstet. 2016;293(2):439–445.
H, Tanaka K, Yamamoto T, Motoyama T, Inoue I, endometriosis with an aromatase inhibitor. Fertil 54. Eskenazi B, Warner ML. Epidemiology of endo-
Enomoto T. Clonal expansion and diversification of Steril. 1998;69(4):709–713. metriosis. Obstet Gynecol Clin North Am. 1997;24(2):
cancer-associated mutations in endometriosis and 40. Attia GR, Zeitoun K, Edwards D, Johns A, Carr BR, 235–258.
normal endometrium. Cell Reports. 2018;24(7): Bulun SE. Progesterone receptor isoform A but not 55. Missmer SA, Cramer DW. The epidemiology of
1777–1789. B is expressed in endometriosis. J Clin Endocrinol endometriosis. Obstet Gynecol Clin North Am. 2003;
26. Nezhat FR, Mahmoud MS. Allen masters peritoneal Metab. 2000;85(8):2897–2902. 30(1):1–19.
defect: a potential pathway to deep infiltrating 41. Kim JJ, Kurita T, Bulun SE. Progesterone action in 56. Viganò P, Parazzini F, Somigliana E, Vercellini P.
rectovaginal endometriosis? J Minim Invasive endometrial cancer, endometriosis, uterine fibroids, Endometriosis: epidemiology and aetiological fac-
Gynecol. 2014;21(3):321–322. and breast cancer. Endocr Rev. 2013;34(1):130–162. tors. Best Pract Res Clin Obstet Gynaecol. 2004;18(2):
27. Brosens I, Pijnenborg R, Benagiano G. Defective 42. Zeitoun K, Takayama K, Sasano H, Suzuki T, 177–200.
myometrial spiral artery remodelling as a cause of Moghrabi N, Andersson S, Johns A, Meng L, Putman 57. Chapron C, Chopin N, Borghese B, Foulot H,
major obstetrical syndromes in endometriosis and M, Carr B, Bulun SE. Deficient 17b-hydroxysteroid Dousset B, Vacher-Lavenu MC, Vieira M, Hasan W,
adenomyosis. Placenta. 2013;34(2):100–105. dehydrogenase type 2 expression in endometriosis: Bricou A. Deeply infiltrating endometriosis: path-
28. Kao LC, Germeyer A, Tulac S, Lobo S, Yang JP, Taylor failure to metabolize 17b-estradiol. J Clin Endocrinol ogenetic implications of the anatomical distribu-
RN, Osteen K, Lessey BA, Giudice LC. Expression Metab. 1998;83(12):4474–4480. tion. Hum Reprod. 2006;21(7):1839–1845.
profiling of endometrium from women with 43. Attar E, Tokunaga H, Imir G, Yilmaz MB, Redwine D, 58. Farquhar C. Endometriosis. BMJ. 2007;334(7587):
endometriosis reveals candidate genes for disease- Putman M, Gurates B, Attar R, Yaegashi N, Hales DB, 249–253.
based implantation failure and infertility. Endocri- Bulun SE. Prostaglandin E2 via steroidogenic factor-1 59. Missmer SA, Hankinson SE, Spiegelman D, Barbieri
nology. 2003;144(7):2870–2881. coordinately regulates transcription of steroidogenic RL, Malspeis S, Willett WC, Hunter DJ. Reproductive
29. Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard genes necessary for estrogen synthesis in endome- history and endometriosis among premenopausal
M, Nezhat CR, Lessey BA, Giudice LC. Gene ex- triosis. J Clin Endocrinol Metab. 2009;94(2):623–631. women. Obstet Gynecol. 2004;104(5 Pt 1):965–974.
pression analysis of endometrium reveals pro- 44. Kettel LM, Murphy AA, Morales AJ, Ulmann A, 60. Thomas EJ. Endometriosis. BMJ. 1993;306(6871):
gesterone resistance and candidate susceptibility Baulieu EE, Yen SS. Treatment of endometriosis 158–159.
genes in women with endometriosis. Endocrinology. with the antiprogesterone mifepristone (RU486). 61. Eaton SB, Pike MC, Short RV, Lee NC, Trussell J,
2007;148(8):3814–3826. Fertil Steril. 1996;65(1):23–28. Hatcher RA, Wood JW, Worthman CM, Jones NG,
30. Tseng JF, Ryan IP, Milam TD, Murai JT, Schriock ED, 45. Gainer EE, Ulmann A. Pharmacologic properties of Konner MJ, Hill KR, Bailey R, Hurtado AM. Women’s
Landers DV, Taylor RN. Interleukin-6 secretion in vitro CDB(VA)-2914. Steroids. 2003;68(10–13):1005–1011. reproductive cancers in evolutionary context. Q Rev
is up-regulated in ectopic and eutopic endometrial 46. Chwalisz K, Perez MC, Demanno D, Winkel C, Biol. 1994;69(3):353–367.
stromal cells from women with endometriosis. J Clin Schubert G, Elger W. Selective progesterone re- 62. Evers J, Dunselman G, Land J, Bouckaert P, Van Der
Endocrinol Metab. 1996;81(3):1118–1122. ceptor modulator development and use in the Linden P. Management of recurrent endometriosis.
31. Hornung D, Klingel K, Dohrn K, Kandolf R, Wallwiener D, treatment of leiomyomata and endometriosis. In: Coutinho EM, Spinola P, Hanson de Moura L,
Taylor RN. Regulated on activation, normal T-cell- Endocr Rev. 2005;26(3):423–438. eds. Progress in the Management of Endometriosis.

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1075


REVIEW

London, UK: Parthenon Publishing Group; 1995: 80. Dmowski WP, Gebel HM, Rawlins RG. Immunologic progesterone in endometriosis. Biol Reprod. 2011;
291–297. aspects of endometriosis. Obstet Gynecol Clin North 84(4):801–815.
63. Vercellini P, Somigliana E, Viganò P, De Matteis S, Am. 1989;16(1):93–103. 96. Lebovic DI, Mueller MD, Taylor RN. Immunobiology
Barbara G, Fedele L. Post-operative endometriosis 81. Osteen KG, Sierra-Rivera E. Does disruption of of endometriosis. Fertil Steril. 2001;75(1):1–10.
recurrence: a plea for prevention based on path- immune and endocrine systems by environmental 97. Chandler RL, Damrauer JS, Raab JR, Schisler JC,
ogenetic, epidemiological and clinical evidence. toxins contribute to development of endometri- Wilkerson MD, Didion JP, Starmer J, Serber D, Yee D,
Reprod Biomed Online. 2010;21(2):259–265. osis? Semin Reprod Endocrinol. 1997;15(3):301–308. Xiong J, Darr DB, Pardo-Manuel de Villena F, Kim
64. Jain S, Dalton ME. Chocolate cysts from ovarian 82. Ota H, Igarashi S, Sasaki M, Tanaka T. Distribution of WY, Magnuson T. Coexistent ARID1A–PIK3CA
follicles. Fertil Steril. 1999;72(5):852–856. cyclooxygenase-2 in eutopic and ectopic endo- mutations promote ovarian clear-cell tumorigen-
65. Scurry J, Whitehead J, Healey M. Classification of metrium in endometriosis and adenomyosis. Hum esis through pro-tumorigenic inflammatory cyto-

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
ovarian endometriotic cysts. Int J Gynecol Pathol. Reprod. 2001;16(3):561–566. kine signalling. Nat Commun. 2015;6(1):6118.
2001;20(2):147–154. 83. Tokushige N, Markham R, Russell P, Fraser IS. High 98. Bulun SE, Simpson ER. Competitive reverse
66. D’Hooghe TM, Debrock S. Endometriosis, retro- density of small nerve fibres in the functional layer transcription-polymerase chain reaction analysis
grade menstruation and peritoneal inflammation in of the endometrium in women with endometriosis. indicates that levels of aromatase cytochrome P450
women and in baboons. Hum Reprod Update. 2002; Hum Reprod. 2006;21(3):782–787. transcripts in adipose tissue of buttocks, thighs, and
8(1):84–88. 84. Barragan F, Irwin JC, Balayan S, Erikson DW, Chen JC, abdomen of women increase with advancing age.
67. Ferguson BR, Bennington JL, Haber SL. Histo- Houshdaran S, Piltonen TT, Spitzer TL, George A, J Clin Endocrinol Metab. 1994;78(2):428–432.
chemistry of mucosubstances and histology of Rabban JT, Nezhat C, Giudice LC. Human endo- 99. Bulun SE, Lin Z, Imir G, Amin S, Demura M, Yilmaz B,
mixed Müllerian pelvic lymph node glandular in- metrial fibroblasts derived from mesenchymal Martin R, Utsunomiya H, Thung S, Gurates B,
clusions. Evidence for histogenesis by Müllerian progenitors inherit progesterone resistance and Tamura M, Langoi D, Deb S. Regulation of aro-
metaplasia of coelomic epithelium. Obstet Gynecol. acquire an inflammatory phenotype in the endo- matase expression in estrogen-responsive breast
1969;33(5):617–625. metrial niche in endometriosis. Biol Reprod. 2016; and uterine disease: from bench to treatment.
68. Sampson JA. Metastatic or embolic endometriosis, 94(5):118. Pharmacol Rev. 2005;57(3):359–383.
due to the menstrual dissemination of endometrial 85. Taylor HS. Endometrial cells derived from donor 100. Ahn SH, Edwards AK, Singh SS, Young SL, Lessey BA,
tissue into the venous circulation. Am J Pathol. 1927; stem cells in bone marrow transplant recipients. Tayade C. IL-17A contributes to the pathogenesis of
3(2): 93–110.43. JAMA. 2004;292(1):81–85. endometriosis by triggering proinflammatory cy-
69. Sasson IE, Taylor HS. Stem cells and the patho- 86. Hufnagel D, Li F, Cosar E, Krikun G, Taylor HS. The tokines and angiogenic growth factors. J Immunol.
genesis of endometriosis. Ann N Y Acad Sci. 2008; role of stem cells in the etiology and pathophysi- 2015;195(6):2591–2600.
1127(1):106–115. ology of endometriosis. Semin Reprod Med. 2015; 101. Greaves E, Collins F, Esnal-Zufiaurre A, Giakoumelou
70. Bulun SE, Monsivais D, Kakinuma T, Furukawa Y, 33(5):333–340. S, Horne AW, Saunders PT. Estrogen receptor (ER)
Bernardi L, Pavone ME, Dyson M. Molecular biology 87. Hapangama DK, Drury J, Da Silva L, Al-Lamee H, agonists differentially regulate neuroangiogenesis in
of endometriosis: from aromatase to genomic ab- Earp A, Valentijn AJ, Edirisinghe DP, Murray PA, peritoneal endometriosis via the repellent factor
Fazleabas AT, Gargett CE. Abnormally located SLIT3. Endocrinology. 2014;155(10):4015–4026.
normalities. Semin Reprod Med. 2015;33(3):220–224.
SSEA11/SOX91 endometrial epithelial cells with a 102. Bulun SE. Endometriosis. In: Strauss J, Barbieri R, eds.
71. Lucidi RS, Witz CA, Chrisco M, Binkley PA, Shain SA,
basalis-like phenotype in the eutopic functionalis Yen & Jaffe’s Reproductive Endocrinology. 8th ed.
Schenken RS. A novel in vitro model of the early
layer may play a role in the pathogenesis of en- Philadelphia, PA: Elsevier; 2019:609–642.
endometriotic lesion demonstrates that attach-
dometriosis. Hum Reprod. 2019;34(1):56–68. 103. Simpson JL, Elias S, Malinak LR, Buttram VC Jr.
ment of endometrial cells to mesothelial cells is
88. Li D, Li H, Wang Y, Eldomany A, Wu J, Yuan C, Xue J, Heritable aspects of endometriosis. I. Genetic
dependent on the source of endometrial cells. Fertil
Shi J, Jia Y, Ha C, Han S, Liu X, Yang J, Liu D. De- studies. Am J Obstet Gynecol. 1980;137(3):327–331.
Steril. 2005;84(1):16–21.
velopment and characterization of a polarized 104. Kennedy S, Mardon H, Barlow D. Familial endo-
72. Giudice LC, Kao LC. Endometriosis. Lancet. 2004;
human endometrial cell epithelia in an air-liquid metriosis. J Assist Reprod Genet. 1995;12(1):32–34.
364(9447):1789–1799.
interface state. Stem Cell Res Ther. 2018;9(1):209. 105. Treloar SA, O’Connor DT, O’Connor VM, Martin
73. Hornung D, Ryan IP, Chao VA, Vigne JL, Schriock ED,
89. Houshdaran S, Nezhat CR, Vo KC, Zelenko Z, Irwin NG. Genetic influences on endometriosis in an
Taylor RN. Immunolocalization and regulation of
JC, Giudice LC. Aberrant endometrial DNA Australian twin sample. Fertil Steril. 1999;71(4):701–
the chemokine RANTES in human endometrial and
methylome and associated gene expression in 710.
endometriosis tissues and cells. J Clin Endocrinol
women with endometriosis. Biol Reprod. 2016;95(5): 106. Montgomery GW, Nyholt DR, Zhao ZZ, Treloar SA,
Metab. 1997;82(5):1621–1628. 93. Painter JN, Missmer SA, Kennedy SH, Zondervan KT.
74. Wu Y, Kajdacsy-Balla A, Strawn E, Basir Z, Halverson 90. Witz CA, Thomas MR, Montoya-Rodriguez IA, Nair The search for genes contributing to endometriosis
G, Jailwala P, Wang Y, Wang X, Ghosh S, Guo SW. AS, Centonze VE, Schenken RS. Short-term culture risk. Hum Reprod Update. 2008;14(5):447–457.
Transcriptional characterizations of differences of peritoneum explants confirms attachment of 107. Rahmioglu N, Missmer SA, Montgomery GW,
between eutopic and ectopic endometrium. En- endometrium to intact peritoneal mesothelium. Zondervan KT. Insights into assessing the genetics
docrinology. 2006;147(1):232–246. Fertil Steril. 2001;75(2):385–390. of endometriosis. Curr Obstet Gynecol Rep. 2012;
75. Osteen KG, Bruner KL, Sharpe-Timms KL. Steroid 91. Meyer R. Zur Frage der heterotopen Epi- 1(3):124–137.
and growth factor regulation of matrix metal- thelwucherung, insbesondere des Peritonealepithels 108. Treloar SA, Zhao ZZ, Le L, Zondervan KT, Martin
loproteinase expression and endometriosis. Semin und in den Ovarien. Virchows Arch. 1924;250(3): NG, Kennedy S, Nyholt DR, Montgomery GW.
Reprod Endocrinol. 1996;14(3):247–255. 595–610. Variants in EMX2 and PTEN do not contribute to
76. Nair AS, Nair HB, Lucidi RS, Kirchner AJ, Schenken RS, 92. Dmowski WP, Ding J, Shen J, Rana N, Fernandez risk of endometriosis. Mol Hum Reprod. 2007;13(8):
Tekmal RR, Witz CA. Modeling the early endo- BB, Braun DP. Apoptosis in endometrial glan- 587–594.
metriotic lesion: mesothelium-endometrial cell co- dular and stromal cells in women with and 109. Zhao ZZ, Nyholt DR, Le L, Treloar SA, Montgomery
culture increases endometrial invasion and alters without endometriosis. Hum Reprod. 2001;16(9): GW. Common variation in the CYP17A1 and IFIT1
mesothelial and endometrial gene transcription. Fertil 1802–1808. genes on chromosome 10 does not contribute to
Steril. 2008;90(4 Suppl):1487–1495. 93. Béliard A, Noël A, Foidart JM. Reduction of apo- the risk of endometriosis. Open Reprod Sci J. 2008;
77. Brosens I, Benagiano G. Is neonatal uterine bleeding ptosis and proliferation in endometriosis. Fertil 1(1):35–40.
involved in the pathogenesis of endometriosis Steril. 2004;82(1):80–85. 110. Lin J, Zong L, Kennedy SH, Zondervan KT. Coding
as a source of stem cells? Fertil Steril. 2013;100(3): 94. Dyson MT, Roqueiro D, Monsivais D, Ercan CM, regions of INHBA, SFRP4 and HOXA10 are not
622–623. Pavone ME, Brooks DC, Kakinuma T, Ono M, Jafari implicated in familial endometriosis linked to
78. Brosens I, Brosens J, Benagiano G. Neonatal uterine N, Dai Y, Bulun SE. Genome-wide DNA methylation chromosome 7p13–15. Mol Hum Reprod. 2011;
bleeding as antecedent of pelvic endometriosis. analysis predicts an epigenetic switch for GATA 17(10):605–611.
Hum Reprod. 2013;28(11):2893–2897. factor expression in endometriosis. PLoS Genet. 111. Zondervan KT, Cardon LR, Kennedy SH. What
79. Gargett CE, Schwab KE, Brosens JJ, Puttemans P, 2014;10(3):e1004158. makes a good case-control study? Design issues for
Benagiano G, Brosens I. Potential role of endo- 95. Aghajanova L, Tatsumi K, Horcajadas JA, Zamah complex traits such as endometriosis. Hum Reprod.
metrial stem/progenitor cells in the pathogenesis of AM, Esteban FJ, Herndon CN, Conti M, Giudice LC. 2002;17(6):1415–1423.
early-onset endometriosis. Mol Hum Reprod. 2014; Unique transcriptome, pathways, and networks in 112. Treloar SA, Wicks J, Nyholt DR, Montgomery GW,
20(7):591–598. the human endometrial fibroblast response to Bahlo M, Smith V, Dawson G, Mackay IJ, Weeks

1076 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

DE, Bennett ST, Carey A, Ewen-White KR, Duffy Endocrinology. 13th ed. Philadelphia, PA: Elsevier; 59 CpG island in endometriosis. J Clin Endocrinol
DL, O’connor DT, Barlow DH, Martin NG, 2016:627–663. Metab. 2007;92(8):3261–3267.
Kennedy SH. Genomewide linkage study in 1,176 122. Hodgkinson K, Forrest LA, Vuong N, Garson K, 134. Utsunomiya H, Cheng YH, Lin Z, Reierstad S, Yin P,
affected sister pair families identifies a significant Djordjevic B, Vanderhyden BC. GREB1 is an estrogen Attar E, Xue Q, Imir G, Thung S, Trukhacheva E,
susceptibility locus for endometriosis on chro- receptor-regulated tumour promoter that is fre- Suzuki T, Sasano H, Kim JJ, Yaegashi N, Bulun SE.
mosome 10q26. Am J Hum Genet. 2005;77(3): quently expressed in ovarian cancer. Oncogene. Upstream stimulatory factor-2 regulates steroido-
365–376. 2018;37(44):5873–5886. genic factor-1 expression in endometriosis. Mol
113. Painter JN, Anderson CA, Nyholt DR, Macgregor S, 123. Veeraraghavan J, Tan Y, Cao XX, Kim JA, Wang X, Endocrinol. 2008;22(4):904–914.
Lin J, Lee SH, Lambert A, Zhao ZZ, Roseman F, Guo Chamness GC, Maiti SN, Cooper LJ, Edwards DP, 135. Ryan IP, Taylor RN. Endometriosis and infertility:
Q, Gordon SD, Wallace L, Henders AK, Visscher PM, Contreras A, Hilsenbeck SG, Chang EC, Schiff R, Wang new concepts. Obstet Gynecol Surv. 1997;52(6):

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
Kraft P, Martin NG, Morris AP, Treloar SA, Kennedy XS. Recurrent ESR1–CCDC170 rearrangements in an 365–371.
SH, Missmer SA, Montgomery GW, Zondervan KT. aggressive subset of oestrogen receptor-positive 136. Tsai SJ, Wu MH, Lin CC, Sun HS, Chen HM. Reg-
Genome-wide association study identifies a locus at breast cancers. Nat Commun. 2014;5(1):4577. ulation of steroidogenic acute regulatory protein
7p15.2 associated with endometriosis. Nat Genet. 124. Doherty JA, Rossing MA, Cushing-Haugen KL, Chen expression and progesterone production in endo-
2011;43(1):51–54. C, Van Den Berg DJ, Wu AH, Pike MC, Ness RB, metriotic stromal cells. J Clin Endocrinol Metab. 2001;
114. Painter JN, Nyholt DR, Morris A, Zhao ZZ, Henders Moysich K, Chenevix-Trench G, Beesley J, Webb PM, 86(12):5765–5773.
AK, Lambert A, Wallace L, Martin NG, Kennedy SH, Chang-Claude J, Wang-Gohrke S, Goodman MT, 137. Sun HS, Hsiao KY, Hsu CC, Wu MH, Tsai SJ.
Treloar SA, Zondervan KT, Montgomery GW. High- Lurie G, Thompson PJ, Carney ME, Hogdall E, Kjaer Transactivation of steroidogenic acute regulatory
density fine-mapping of a chromosome 10q26 SK, Hogdall C, Goode EL, Cunningham JM, Fridley protein in human endometriotic stromalcells is
linkage peak suggests association between endo- BL, Vierkant RA, Berchuck A, Moorman PG, mediated by the prostaglandin EP2 receptor. En-
metriosis and variants close to CYP2C19. Fertil Steril. Schildkraut JM, Palmieri RT, Cramer DW, Terry KL, docrinology. 2003;144(9):3934–3942.
2011;95(7):2236–2240. Yang HP, Garcia-Closas M, Chanock S, Lissowska J, 138. Bulun SE, Yang S, Fang Z, Gurates B, Tamura M,
115. Mwinyi J, Cavaco I, Pedersen RS, Persson A, Song H, Pharoah PD, Shah M, Perkins B, McGuire V, Zhou J, Sebastian S. Role of aromatase in endo-
Burkhardt S, Mkrtchian S, Ingelman-Sundberg M. Whittemore AS, Di Cioccio RA, Gentry-Maharaj A, metrial disease. J Steroid Biochem Mol Biol. 2001;
Regulation of CYP2C19 expression by estrogen Menon U, Gayther SA, Ramus SJ, Ziogas A, Brewster 79(1-5):19–25.
receptor a: implications for estrogen-dependent W, Anton-Culver H, Pearce CL; Australian Ovarian 139. Olive DL, Pritts EA. Treatment of endometriosis.
inhibition of drug metabolism. Mol Pharmacol. Cancer Study Management Group; Australian N Engl J Med. 2001;345(4):266–275.
2010;78(5):886–894. Cancer Study (Ovarian Cancer); Ovarian Cancer 140. Zeitoun KM, Bulun SE. Aromatase: a key molecule
116. Zondervan KT, Treloar SA, Lin J, Weeks DE, Nyholt Association Consortium (OCAC). ESR1/SYNE1 in the pathophysiology of endometriosis and a
DR, Mangion J, MacKay IJ, Cardon LR, Martin NG, polymorphism and invasive epithelial ovarian cancer therapeutic target. Fertil Steril. 1999;72(6):961–969.
Kennedy SH, Montgomery GW. Significant evi- risk: an Ovarian Cancer Association Consortium 141. Attar E, Bulun SE. Aromatase inhibitors: the next
study. Cancer Epidemiol Biomarkers Prev. 2010;19(1): generation of therapeutics for endometriosis? Fertil
dence of one or more susceptibility loci for en-
245–250.
dometriosis with near-Mendelian inheritance on Steril. 2006;85(5):1307–1318.
125. Grechukhina O, Petracco R, Popkhadze S, Massasa E,
chromosome 7p13–15. Hum Reprod. 2007;22(3): 142. Xue Q, Lin Z, Cheng YH, Huang CC, Marsh E, Yin P,
Paranjape T, Chan E, Flores I, Weidhaas JB, Taylor HS.
717–728. Milad MP, Confino E, Reierstad S, Innes J, Bulun SE.
A polymorphism in a let-7 microRNA binding site of
117. Uno S, Zembutsu H, Hirasawa A, Takahashi A, Kubo Promoter methylation regulates estrogen receptor
KRAS in women with endometriosis. EMBO Mol
M, Akahane T, Aoki D, Kamatani N, Hirata K, 2 in human endometrium and endometriosis. Biol
Med. 2012;4(3):206–217.
Nakamura Y. A genome-wide association study Reprod. 2007;77(4):681–687.
126. Dyson MT, Kakinuma T, Pavone ME, Monsivais
identifies genetic variants in the CDKN2BAS locus 143. Juhasz-Böss I, Fischer C, Lattrich C, Skrzypczak M,
D, Navarro A, Malpani SS, Ono M, Bulun SE.
associated with endometriosis in Japanese. Nat Malik E, Ortmann O, Treeck O. Endometrial ex-
Aberrant expression and localization of deoxy-
Genet. 2010;42(8):707–710. pression of estrogen receptor b and its splice
ribonucleic acid methyltransferase 3B in endometri-
118. Nyholt DR, Low SK, Anderson CA, Painter JN, Uno S, variants in patients with and without endometri-
otic stromal cells. Fertil Steril. 2015;104(4):953–963.e2.
Morris AP, MacGregor S, Gordon SD, Henders AK, osis. Arch Gynecol Obstet. 2011;284(4):885–891.
127. Bernardi LA, Dyson MT, Tokunaga H, Sison C, Oral
Martin NG, Attia J, Holliday EG, McEvoy M, Scott RJ, 144. Han SJ, Hawkins SM, Begum K, Jung SY, Kovanci E,
M, Robins JC, Bulun SE. The essential role of GATA6
Kennedy SH, Treloar SA, Missmer SA, Adachi S, Qin J, Lydon JP, DeMayo FJ, O’Malley BW. A new
in the activation of estrogen synthesis in endo-
Tanaka K, Nakamura Y, Zondervan KT, Zembutsu metriosis. Reprod Sci. 2019;26(1):60–69. isoform of steroid receptor coactivator-1 is crucial
H, Montgomery GW. Genome-wide association 128. Naqvi H, Ilagan Y, Krikun G, Taylor HS. Altered for pathogenic progression of endometriosis. Nat
meta-analysis identifies new endometriosis risk loci. genome-wide methylation in endometriosis. Reprod Med. 2012;18(7):1102–1111.
Nat Genet. 2012;44(12):1355–1359. Sci. 2014;21(10):1237–1243. 145. Trukhacheva E, Lin Z, Reierstad S, Cheng YH, Milad
119. Rahmioglu N, Nyholt DR, Morris AP, Missmer SA, 129. Lee B, Du H, Taylor HS. Experimental murine en- M, Bulun SE. Estrogen receptor (ER) b regulates ERa
Montgomery GW, Zondervan KT. Genetic variants dometriosis induces DNA methylation and altered expression in stromal cells derived from ovarian
underlying risk of endometriosis: insights from gene expression in eutopic endometrium. Biol endometriosis. J Clin Endocrinol Metab. 2009;94(2):
meta-analysis of eight genome-wide association Reprod. 2009;80(1):79–85. 615–622.
and replication datasets. Hum Reprod Update. 2014; 130. Rubel CA, Wu SP, Lin L, Wang T, Lanz RB, Li X, 146. Tamura M, Deb S, Sebastian S, Okamura K, Bulun
20(5):702–716. Kommagani R, Franco HL, Camper SA, Tong Q, SE. Estrogen up-regulates cyclooxygenase-2 via
120. Sapkota Y, Steinthorsdottir V, Morris AP, Jeong JW, Lydon JP, DeMayo FJ. A Gata2-dependent estrogen receptor in human uterine microvascular
Fassbender A, Rahmioglu N, De Vivo I, Buring JE, transcription network regulates uterine progesterone endothelial cells. Fertil Steril. 2004;81(5):1351–
Zhang F, Edwards TL, Jones S, O D, Peterse D, responsiveness and endometrial function. Cell Re- 1356.
Rexrode KM, Ridker PM, Schork AJ, MacGregor S, ports. 2016;17(5):1414–1425. 147. Daniels S, Robbins J, West CR, Nemeth MA. Cele-
Martin NG, Becker CM, Adachi S, Yoshihara K, 131. Lala DS, Rice DA, Parker KL. Steroidogenic factor I, a coxib in the treatment of primary dysmenorrhea:
Enomoto T, Takahashi A, Kamatani Y, Matsuda K, key regulator of steroidogenic enzyme expression, is results from two randomized, double-blind, active-
Kubo M, Thorleifsson G, Geirsson RT, the mouse homolog of fushi tarazu-factor I. Mol and placebo-controlled, crossover studies. Clin Ther.
Thorsteinsdottir U, Wallace LM, Yang J, Velez Endocrinol. 1992;6(8):1249–1258. 2009;31(6):1192–1208.
Edwards DR, Nyegaard M, Low SK, Zondervan 132. Zeitoun K, Takayama K, Michael MD, Bulun SE. 148. Vercellini P, Cortesi I, Crosignani PG. Progestins for
KT, Missmer SA, D’Hooghe T, Montgomery GW, Stimulation of aromatase P450 promoter (II) activity symptomatic endometriosis: a critical analysis of the
Chasman DI, Stefansson K, Tung JY, Nyholt DR; in endometriosis and its inhibition in endometrium evidence. Fertil Steril. 1997;68(3):393–401.
iPSYCH-SSI-Broad Group. Meta-analysis identifies are regulated by competitive binding of steroidogenic 149. Surrey ES. The role of progestins in treating the pain
five novel loci associated with endometriosis factor-1 and chicken ovalbumin upstream promoter of endometriosis. J Minim Invasive Gynecol. 2006;
highlighting key genes involved in hormone transcription factor to the same cis-acting element. 13(6):528–534.
metabolism. Nat Commun. 2017;8:15539. Mol Endocrinol. 1999;13(2):239–253. 150. Madauss KP, Grygielko ET, Deng SJ, Sulpizio AC,
121. Bulun SE. Physiology and pathology of the female 133. Xue Q, Lin Z, Yin P, Milad MP, Cheng YH, Confino E, Stanley TB, Wu C, Short SA, Thompson SK, Stewart
reproductive axis. In: Melmed S, Polonsky K, Larsen Reierstad S, Bulun SE. Transcriptional activation of EL, Laping NJ, Williams SP, Bray JD. A structural and
P, Kronenberg H, eds. Williams Textbook of steroidogenic factor-1 by hypomethylation of the in vitro characterization of asoprisnil: a selective

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1077


REVIEW

progesterone receptor modulator. Mol Endocrinol. Global gene profiling in human endometrium 179. Coleman RE, Marshall H, Cameron D, Dodwell D,
2007;21(5):1066–1081. during the window of implantation. Endocrinology. Burkinshaw R, Keane M, Gil M, Houston SJ, Grieve
151. Jackson KS, Brudney A, Hastings JM, Mavrogianis 2002;143(6):2119–2138. RJ, Barrett-Lee PJ, Ritchie D, Pugh J, Gaunt C, Rea U,
PA, Kim JJ, Fazleabas AT. The altered distribution of 165. Bulun S, Adashi E. The physiology and pathology of Peterson J, Davies C, Hiley V, Gregory W, Bell R;
the steroid hormone receptors and the chaperone the female reproductive axis. In: Kronenberg HM, AZURE Investigators. Breast-cancer adjuvant ther-
immunophilin FKBP52 in a baboon model of en- Melmed S, Polonsky KS, Larsen PR, eds. Williams apy with zoledronic acid. N Engl J Med. 2011;365(15):
dometriosis is associated with progesterone re- Textbook of Endocrinology, 10th ed. Philadelphia, 1396–1405.
sistance during the window of uterine receptivity. PA: Elsevier Health Sciences; 2003:587–663. 180. Kettel LM, Murphy AA, Morales AJ, Yen SS. Pre-
Reprod Sci. 2007;14(2):137–150. 166. Brosens JJ, Hayashi N, White JO. Progesterone re- liminary report on the treatment of endometriosis
152. Tseng L, Gurpide E. Estradiol and 20a-dihy- ceptor regulates decidual prolactin expression in with low-dose mifepristone (RU 486). Am J Obstet

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
droprogesterone dehydrogenase activities in human differentiating human endometrial stromal cells. Gynecol. 1998;178(6):1151–1156.
endometrium during the menstrual cycle. Endocri- Endocrinology. 1999;140(10):4809–4820. 181. Donnez J, Tomaszewski J, Vázquez F, Bouchard P,
nology. 1974;94(2):419–423. 167. Fazleabas AT, Brudney A, Chai D, Langoi D, Bulun SE. Lemieszczuk B, Baró F, Nouri K, Selvaggi L, Sodowski
153. Tseng L, Gurpide E. Induction of human endo- Steroid receptor and aromatase expression in ba- K, Bestel E, Terrill P, Osterloh I, Loumaye E; PEARL II
metrial estradiol dehydrogenase by progestins. En- boon endometriotic lesions. Fertil Steril. 2003; Study Group. Ulipristal acetate versus leuprolide
docrinology. 1975;97(4):825–833. 80(Suppl 2):820–827. acetate for uterine fibroids. N Engl J Med. 2012;
154. Satyaswaroop PG, Wartell DJ, Mortel R. Dis- 168. Osteen KG, Bruner-Tran KL, Eisenberg E. Reduced 366(5):421–432.
tribution of progesterone receptor, estradiol progesterone action during endometrial matura- 182. Chabbert-Buffet N, Pintiaux-Kairis A, Bouchard P;
dehydrogenase, and 20a-dihydroprogesterone de- tion: a potential risk factor for the development of VA2914 Study Group. Effects of the progesterone
hydrogenase activities in human endometrial endometriosis. Fertil Steril. 2005;83(3):529–537. receptor modulator VA2914 in a continuous low
glands and stroma: progestin induction of steroid 169. Dyson MT, Bulun SE. Cutting SRC-1 down to size in dose on the hypothalamic-pituitary-ovarian axis
dehydrogenase activities in vitro is restricted to the endometriosis. Nat Med. 2012;18(7):1016–1018. and endometrium in normal women: a prospective,
glandular epithelium. Endocrinology. 1982;111(3): 170. Bedaiwy MAA, Allaire C, Yong P, Alfaraj S. Medical randomized, placebo-controlled trial. J Clin Endo-
743–749. management of endometriosis in patients with chronic crinol Metab. 2007;92(9):3582–3589.
155. Yang S, Fang Z, Gurates B, Tamura M, Miller J, Ferrer pelvic pain. Semin Reprod Med. 2017;35(1):38–53. 183. Donnez J, Arriagada P, Marciniak M, Larrey D. Liver
K, Bulun SE. Stromal PRs mediate induction of 17b- 171. Barbieri RL. Hormone treatment of endometriosis: safety parameters of ulipristal acetate for the
hydroxysteroid dehydrogenase type 2 expression the estrogen threshold hypothesis. Am J Obstet treatment of uterine fibroids: a comprehensive
in human endometrial epithelium: a paracrine Gynecol. 1992;166(2):740–745. review of the clinical development program. Expert
mechanism for inactivation of E2. Mol Endocrinol. 172. Carr B, Dmowski WP, O’Brien C, Jiang P, Burke J, Opin Drug Saf. 2018;17(12):1225–1232.
2001;15(12):2093–2105. Jimenez R, Garner E, Chwalisz K. Elagolix, an oral 184. Jabbour HN, Sales KJ, Smith OP, Battersby S, Boddy
156. Casey ML, MacDonald PC, Andersson S. 17 beta- GnRH antagonist, versus subcutaneous depot SC. Prostaglandin receptors are mediators of vas-
Hydroxysteroid dehydrogenase type 2: chromo- medroxyprogesterone acetate for the treatment of cular function in endometrial pathologies. Mol Cell
somal assignment and progestin regulation of gene endometriosis: effects on bone mineral density. Endocrinol. 2006;252(1–2):191–200.
expression in human endometrium. J Clin Invest. Reprod Sci. 2014;21(11):1341–1351. 185. Hayes EC, Rock JA. COX-2 inhibitors and their role in
1994;94(5):2135–2141. 173. Taylor HS, Giudice LC, Lessey BA, Abrao MS, gynecology. Obstet Gynecol Surv. 2002;57(11):768–780.
157. Mustonen MV, Isomaa VV, Vaskivuo T, Tapanainen Kotarski J, Archer DF, Diamond MP, Surrey E, 186. Funk CD. Prostaglandins and leukotrienes: advances
J, Poutanen MH, Stenbäck F, Vihko RK, Vihko PT. Johnson NP, Watts NB, Gallagher JC, Simon JA, Carr in eicosanoid biology. Science. 2001;294(5548):
Human 17b-hydroxysteroid dehydrogenase type 2 1871–1875.
BR, Dmowski WP, Leyland N, Rowan JP, Duan WR,
messenger ribonucleic acid expression and locali- 187. Mancini JA, Blood K, Guay J, Gordon R, Claveau D,
Ng J, Schwefel B, Thomas JW, Jain RI, Chwalisz K.
zation in term placenta and in endometrium during Chan CC, Riendeau D. Cloning, expression, and up-
Treatment of endometriosis-associated pain with
the menstrual cycle. J Clin Endocrinol Metab. 1998; regulation of inducible rat prostaglandin e synthase
elagolix, an oral GnRH antagonist. N Engl J Med.
83(4):1319–1324. during lipopolysaccharide-induced pyresis and
2017;377(1):28–40.
158. Cheng YH, Yin P, Xue Q, Yilmaz B, Dawson MI, adjuvant-induced arthritis. J Biol Chem. 2001;276(6):
174. Struthers RS, Nicholls AJ, Grundy J, Chen T, Jimenez
Bulun SE. Retinoic acid (RA) regulates 17b- 4469–4475.
R, Yen SS, Bozigian HP. Suppression of gonado-
hydroxysteroid dehydrogenase type 2 expression in 188. Wu MH, Wang CA, Lin CC, Chen LC, Chang WC,
tropins and estradiol in premenopausal women
endometrium: interaction of RA receptors with Tsai SJ. Distinct regulation of cyclooxygenase-2 by
by oral administration of the nonpeptide gonadotropin-
specificity protein (SP) 1/SP3 for estradiol metab- interleukin-1b in normal and endometriotic stromal
olism. J Clin Endocrinol Metab. 2008;93(5):1915– releasing hormone antagonist elagolix. J Clin Endocrinol cells. J Clin Endocrinol Metab. 2005;90(1):286–295.
1923. Metab. 2009;94(2):545–551. 189. Ni H, Sun T, Ding NZ, Ma XH, Yang ZM. Differential
159. Cheng YH, Imir A, Fenkci V, Yilmaz MB, Bulun SE. 175. Surrey E, Taylor HS, Giudice L, Lessey BA, Abrao MS, expression of microsomal prostaglandin e synthase
Stromal cells of endometriosis fail to produce paracrine Archer DF, Diamond MP, Johnson NP, Watts NB, at implantation sites and in decidual cells of mouse
factors that induce epithelial 17b-hydroxysteroid de- Gallagher JC, Simon JA, Carr BR, Dmowski WP, uterus. Biol Reprod. 2002;67(1):351–358.
hydrogenase type 2 gene and its transcriptional reg- Leyland N, Singh SS, Rechberger T, Agarwal SK, 190. Sun T, Li SJ, Diao HL, Teng CB, Wang HB, Yang ZM.
ulator Sp1: a mechanism for defective estradiol Duan WR, Schwefel B, Thomas JW, Peloso PM, Ng J, Cyclooxygenases and prostaglandin E synthases
metabolism. Am J Obstet Gynecol. 2007;196(4):391.e1–7. Soliman AM, Chwalisz K. Long-term outcomes of in the endometrium of the rhesus monkey dur-
160. Bulun SE, Cheng YH, Yin P, Imir G, Utsunomiya H, elagolix in women with endometriosis: results from ing the menstrual cycle. Reproduction. 2004;127(4):
Attar E, Innes J, Julie Kim J. Progesterone resistance two extension studies. Obstet Gynecol. 2018;132(1): 465–473.
in endometriosis: link to failure to metabolize es- 147–160. 191. Jabbour HN, Milne SA, Williams AR, Anderson RA,
tradiol. Mol Cell Endocrinol. 2006;248(1–2):94–103. 176. Melis GB, Neri M, Corda V, Malune ME, Piras B, Boddy SC. Expression of COX-2 and PGE synthase
161. Pavone ME, Reierstad S, Sun H, Milad M, Bulun SE, Pirarba S, Guerriero S, Orrù M, D’Alterio MN, and synthesis of PGE2 in endometrial adenocarci-
Cheng YH. Altered retinoid uptake and action Angioni S, Paoletti AM. Overview of elagolix for the noma: a possible autocrine/paracrine regulation of
contributes to cell survival in endometriosis. J Clin treatment of endometriosis. Expert Opin Drug neoplastic cell function via EP2/EP4 receptors. Br J
Endocrinol Metab. 2010;95(11):E300–E309. Metab Toxicol. 2016;12(5):581–588. Cancer. 2001;85(7):1023–1031.
162. Pavone ME, Dyson M, Reirstad S, Pearson E, Ishikawa 177. Committee opinion no. 663: aromatase inhibitors in 192. Tamura M, Sebastian S, Yang S, Gurates B, Fang Z,
H, Cheng YH, Bulun SE. Endometriosis expresses a gynecologic practice. Obstet Gynecol. 2016;127(6): Bulun SE. Interleukin-1beta elevates cyclooxygenase-2
molecular pattern consistent with decreased reti- e170–e174. protein level and enzyme activity via increasing its
noid uptake, metabolism and action. Hum Reprod. 178. Chlebowski RT, Rohan TE, Manson JE, Aragaki AK, mRNA stability in human endometrial stromal cells:
2011;26(8):2157–2164. Kaunitz A, Stefanick ML, Simon MS, Johnson KC, an effect mediated by extracellularly regulated ki-
163. Schug TT, Berry DC, Shaw NS, Travis SN, Noy N. Wactawski-Wende J, O’Sullivan MJ, Adams-Campbell nases 1 and 2. J Clin Endocrinol Metab. 2002;87(7):
Opposing effects of retinoic acid on cell growth LL, Nassir R, Lessin LS, Prentice RL. Breast cancer after 3263–3273.
result from alternate activation of two different use of estrogen plus progestin and estrogen alone: 193. Tamura M, Sebastian S, Yang S, Gurates B, Ferrer K,
nuclear receptors. Cell. 2007;129(4):723–733. analyses of data from 2 Women’s Health Initiative Sasano H, Okamura K, Bulun SE. Up-regulation of
164. Kao LC, Tulac S, Lobo S, Imani B, Yang JP, Germeyer randomized clinical trials. JAMA Oncol. 2015;1(3): cyclooxygenase-2 expression and prostaglandin
A, Osteen K, Taylor RN, Lessey BA, Giudice LC. 296–305. synthesis in endometrial stromal cells by malignant

1078 Bulun et al Endometriosis Endocrine Reviews, August 2019, 40(4):1048–1079


REVIEW

endometrial epithelial cells. A paracrine effect extent of induced endometriosis in the baboon. 208. Jin RU, Mills JC. Are gastric and esophageal meta-
mediated by prostaglandin E2 and nuclear factor- Hum Reprod. 2006;21(7):1856–1862. plasia relatives? The case for Barrett’s stemming
kB. J Biol Chem. 2002;277(29):26208–26216. 200. Benaglia L, Somigliana E, Vighi V, Ragni G, Vercellini from SPEM. Dig Dis Sci. 2018;63(8):2028–2041.
194. Tamura M, Sebastian S, Gurates B, Yang S, Fang Z, P, Fedele L. Rate of severe ovarian damage following
Bulun SE. Vascular endothelial growth factor up- surgery for endometriomas. Hum Reprod. 2010;
regulates cyclooxygenase-2 expression in human 25(3):678–682. Acknowledgments
endothelial cells. J Clin Endocrinol Metab. 2002;87(7): 201. Hamdan M, Omar SZ, Dunselman G, Cheong Y. Financial Support: This work was supported by National
3504–3507. Influence of endometriosis on assisted re- Institutes of Health/National Institute of Child Health and
195. Arosh JA, Lee J, Balasubbramanian D, Stanley JA, productive technology outcomes: a systematic Human Development Grant R37-HD38691 (to S.E.B.).
Long CR, Meagher MW, Osteen KG, Bruner-Tran review and meta-analysis. Obstet Gynecol. 2015; Correspondence and Reprint Requests: Serdar E.

Downloaded from https://academic.oup.com/edrv/article-abstract/40/4/1048/5469279 by Galter Health Sciences Library user on 29 July 2019
KL, Burghardt RC, Starzinski-Powitz A, Banu SK. 125(1):79–88. Bulun, MD, Prentice Women’s Hospital, 250 East Superior
Molecular and preclinical basis to inhibit PGE2 202. D’Hooghe TM, Denys B, Spiessens C, Meuleman C, Street, Room 3-2306, Chicago, Illinois 60611. E-mail: s-bulun@
receptors EP2 and EP4 as a novel nonsteroidal Debrock S. Is the endometriosis recurrence rate northwestern.edu.
increased after ovarian hyperstimulation? Fertil Disclosure Summary: The authors have nothing to
therapy for endometriosis. Proc Natl Acad Sci USA.
Steril. 2006;86(2):283–290. disclose.
2015;112(31):9716–9721.
203. Harb HM, Gallos ID, Chu J, Harb M,
196. Reis FM, Petraglia F, Taylor RN. Endometriosis:
Coomarasamy A. The effect of endometriosis on
hormone regulation and clinical consequences of Abbreviations
in vitro fertilisation outcome: a systematic re-
chemotaxis and apoptosis. Hum Reprod Update. CCDC170, coiled-coil domain containing 170; COX, cyclo-
view and meta-analysis. BJOG. 2013;120(11):
2013;19(4):406–418. oxygenase; CpG, cytosine-phosphate-guanine dinucleotide,
1308–1320. CYP17A1; 17-hydroxylase/17,20-lyase, CYP19A1; aromatase,
197. Akoum A, Lemay A, Maheux R. Estradiol and in-
204. de Ziegler D, Pirtea P, Galliano D, Cicinelli E, DNMT; DNA methyltransferase, ER; estrogen receptor, ESR1;
terleukin-1b exert a synergistic stimulatory effect
Meldrum D. Optimal uterine anatomy and ERa, ESR2; ERb, FSHB; FSH b-subunit, GATA2; GATA-binding
on the expression of the chemokine regulated upon
physiology necessary for normal implantation factor-2, GATA6; GATA-binding factor-6, GREB1; growth
activation, normal T cell expressed, and secreted in
and placentation. Fertil Steril. 2016;105(4):844– regulation by estrogen in breast cancer-1, GWAS; genome-
endometriotic cells. J Clin Endocrinol Metab. 2002; 854. wide association study, HSD3B2; 3b-hydroxysteroid
87(12):5785–5792. 205. Daley GQ. The promise and perils of stem cell dehydrogenase-2, HSD17B2; 17b-hydroxysteroid dehydro-
198. D’Hooghe TM, Nugent NP, Cuneo S, Chai DC, therapeutics. Cell Stem Cell. 2012;10(6):740– genase type 2, iPS; induced pluripotent stem, IVD; in vitro
Deer F, Debrock S, Kyama CM, Mihalyi A, Mwenda 749. decidualization, IVF; in vitro fertilization, mPGES; microsomal
JM. Recombinant human TNFRSF1A (r-hTBP1) 206. Mutlu L, Hufnagel D, Taylor HS. The endometrium PGE synthase, PGE2; prostaglandin E2, PGF; prostaglandin F,
inhibits the development of endometriosis in as a source of mesenchymal stem cells for re- PGH2; prostaglandin H2, PPAR; peroxisome proliferator-
baboons: a prospective, randomized, placebo- and generative medicine. Biol Reprod. 2015;92(6):138. activated receptor, PR; progesterone receptor, RA; retinoic
drug-controlled study. Biol Reprod. 2006;74(1): 207. Miyazaki K, Dyson MT, Coon V JS, Furukawa Y, acid, RAR; RA receptor, RERG; Ras-like, estrogen-regulated;
131–136. Yilmaz BD, Maruyama T, Bulun SE. Generation of growth inhibitor, SF1; steroidogenic factor-1, SNP; single-
199. Falconer H, Mwenda JM, Chai DC, Wagner C, Song progesterone-responsive endometrial stromal fi- nucleotide polymorphism, SRC; steroid receptor coac-
XY, Mihalyi A, Simsa P, Kyama C, Cornillie FJ, broblasts from human induced pluripotent stem tivator, STAR; steroidogenic acute regulatory protein, SYNE1;
Bergqvist A, Fried G, D’Hooghe TM. Treatment with cells: role of the WNT/CTNNB1 pathway. Stem spectrin repeat containing, nuclear envelope-1; UPA, uli-
anti-TNF monoclonal antibody (c5N) reduces the Cell Reports. 2018;11(5):1136–1155. pristal acetate.

doi: 10.1210/er.2018-00242 https://academic.oup.com/edrv 1079


View publication stats

You might also like