Вы находитесь на странице: 1из 12

The

n e w e ng l a n d j o u r na l

of

m e dic i n e

review article
Mechanisms of Disease

Endometriosis
Serdar E. Bulun, M.D.
From the Division of Reproductive Biology Research, Department of Obstetrics
and Gynecology, Feinberg School of
Medicine, Northwestern University, Chicago. Address reprint requests to Dr. Bulun at the Division of Reproductive Biology Research, Dept. of Obstetrics and
Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior St., Rm. 4-123, Chicago, IL 60611,
or at s-bulun@northwestern.edu.
N Engl J Med 2009;360:268-79.
Copyright 2009 Massachusetts Medical Society.

268

ndometriosis is an estrogen-dependent inflammatory disease


that affects 5 to 10% of women of reproductive age in the United States.1 Its
defining feature is the presence of endometrium-like tissue in sites outside
the uterine cavity, primarily on the pelvic peritoneum and ovaries. The main clinical
features are chronic pelvic pain, pain during intercourse, and infertility.1 Endometriosis can be the result of diverse anatomical or biochemical aberrations of uterine
function. For example, endometriosis commonly develops in young women with vaginal obstruction of outflow, possibly because of large quantities of backwashed menstrual tissue that has become implanted on pelvic organs.2 In contrast, endometriosis can also involve mechanisms that are independent of anatomical abnormalities;
for example, the incidence of endometriosis is increased in women who were exposed in utero to environmental toxins or potent estrogens such as diethylstilbestrol.3 As cellular and molecular mechanisms involved in endometriosis are being
uncovered, the diseases classification is evolving from a local disorder to a complex,
chronic systemic disease. Endometriosis can be inherited in a polygenic manner; its
incidence in relatives of affected women is up to seven times the incidence in women
without such a family history.4 There is evidence of linkage to chromosomes 7 and
10, but no relevant genes in these regions appear to have yet been identified.5,6
The three clinically distinct forms of endometriosis are endometriotic implants
on the surface of the pelvic peritoneum and ovaries (peritoneal endometriosis), ovarian cysts lined by endometrioid mucosa (endometriomas), and a complex solid mass
comprised of endometriotic tissue blended with adipose and fibromuscular tissue,
residing between the rectum and the vagina (rectovaginal endometriotic nodule). All
three types may be variants of the same pathologic process or they can be caused
by different mechanisms.7,8 Their common histologic features are the presence of
endometrial stromal or epithelial cells, chronic bleeding, and signs of inflammation. These lesions can occur singly or in combination and are associated with an
increased risk of infertility or chronic pelvic pain.9,10 The inflammation involved in
endometriosis can stimulate nerve endings in the pelvis and thereby cause pain,
impair the function of uterine tubes, decrease receptivity of the endometrium, and
hinder development of the oocyte and embryo.11,12 Endometriosis can also cause
infertility by physically blocking the fallopian tubes.9,10
The treatment of infertility caused by endometriosis is surgical removal of endometriotic tissue or assisted reproductive technology, whereas the usual treatment of
pain is a combination of medical suppression of ovulation and surgery. Peritoneal
implants are resected or vaporized by means of an electric current or laser. Ovarian
endometriomas and rectovaginal endometriotic nodules, however, can be removed
effectively only with the use of full dissection. Epidemiologic and laboratory data suggest a link between ovarian endometriosis and distinct types of ovarian cancer.13,14
Clinical evidence clearly points to a deleterious effect of uninterrupted ovulatory
cycles on the development and persistence of endometriosis.15,16 First, symptoms
of endometriosis usually appear after menarche and vanish after menopause.17 Occan engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

Mechanisms of Disease

sionally, a rectovaginal nodule remains symptomatic in a postmenopausal woman, suggesting that


its persistence is independent of ovarian estrogen
secretion.18 Second, multiparity is associated with
a decreased risk of endometriosis.17 Third, the
disruption of ovulation by analogues of gonadotropin-releasing hormone (GnRH), oral contraceptives, or progestins reduces pelvic disease and
the associated pain.16
In line with these clinical observations are the
findings that indicate major roles of the ovarian
steroids estrogen and progesterone in the development of endometriosis. In humans and other
primates, estrogen stimulates the growth of endometriotic tissue, whereas aromatase inhibitors
that block estrogen formation are beneficial, as
are antiprogestins, in patients with endometriosis.19-21 Levels of nuclear receptors for estrogen
and progesterone are strikingly perturbed in endometriotic tissue as compared with normal endometrium.22-24 Finally, biologically significant quantities of progesterone and estrogen are produced
locally in endometriotic tissue, through an abnormally active steroidogenic cascade that includes
aromatase.25 This review deals mainly with steroid-related mechanisms of endometriosis.

Cel lul a r Or igins


There is no consensus concerning the histologic
origin of endometriosis. Sampson proposed that
fragments of menstrual endometrium pass backward through the fallopian tubes and then become implanted on peritoneal surfaces and persist there.26 This mechanism was shown in primate
models, was observed in humans, and is supported by the observation that endometriosis occurs
exclusively in species that menstruate (i.e., humans
and other primates).27 In contrast, the coelomicmetaplasia hypothesis proposes that the genesis
of endometriotic lesions within the peritoneal cavity is the differentiation of mesothelial cells into
endometrium-like tissue.28 A third hypothesis argues that menstrual tissue from the endometrial
cavity travels to other sites through veins or lymphatic vessels.29 Another proposal is that circulating blood cells originating from bone marrow
can differentiate into endometriotic tissue at various sites.30 Proving or disproving these hypotheses is challenging because of the difficulty in constructing clinically relevant models.
Sampsons implantation hypothesis is a plausible mechanism for most endometriotic lesions

but does not explain why endometriosis develops


in some, but not most, women. Most women have
reflux menstruation into the peritoneal cavity, but
endometriosis occurs in only 5 to 10%. One of two
mechanisms could explain the successful implantation of refluxed endometrium onto the peritoneal surface: molecular defects or immunologic
abnormalities (or both).31 In endometriosis, the
eutopic endometrium exhibits multiple subtle but
biologically important molecular abnormalities,
including the activation of oncogenic pathways
(e.g., the Wingless-type MMTV integration site
family member Wnt or the rat sarcoma viral oncogene homologue Ras) or biosynthetic cascades
favoring increased production of estrogen, cyto
kines, prostaglandins, and metalloproteinases.1,32-36
When the eutopic endometrium, biologically distinct tissue, attaches to mesothelial cells, the magnitude of the molecular abnormalities is amplified drastically, enhancing the survival of the
implant.37 A possible second mechanism of implant survival entails a failure of the immune system to clear implants from the peritoneal surface.1,38,39 Both mechanisms may contribute to the
development of endometriosis.

Mol ecul a r Mech a nisms


There are clear molecular distinctions between endometriotic tissue and endometrium, such as the
overproduction of estrogen, prostaglandins, and
cytokines in endometriotic tissue (Fig. 1).32,40,41
Subtle forms of these abnormalities also occur in
endometrium from a woman with endometriosis
as compared with endometrium from a woman
without the disease (Fig. 1).32,40,41 Moreover, geneexpression profiling of endometrium from women
with endometriosis as compared with endometrium from disease-free women has revealed candidate genes related to implantation failure, infertility, and progesterone resistance.35,42
Inflammation, a feature of endometriotic tissue, is associated with the overproduction of prostaglandins, metalloproteinases, cytokines, and
chemokines.1,32-36,43 Increased levels of acute inflammatory cytokines such as interleukin-1, interleukin-6, and tumor necrosis factor probably
enhance the adhesion of shed endometrial-tissue
fragments onto peritoneal surfaces, and proteolytic membrane metalloproteinases may further
promote implantation of the fragments.1,32-36,43
Monocyte chemoattractant protein 1, interleukin-8,
and RANTES (regulated upon activation normal

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

269

The

n e w e ng l a n d j o u r na l

of

m e dic i n e

A Endometrium in disease-free women


Precursor
Precursor
Estradiol

COX-2

Aromatase
ma
HSD17B2

Low PGE2

Estrogen
trog

Low PGE2 locally

Estrone

B Endometrium in women with endometriosis


Precursor
Precursor
Estradiol

COX-2
Aromatase
HSD17B2

Moderate PGE2
Moderate
PGE2, and low
estradiol, locally

Estrogen
Estrone

C Ectopic endometriotic tissue


Precursor
Precursor
Estradiol

COX-2
Aromatase

HSD17B2
D1

High PGE2
High PGE2
and estradiol
locally

High estrogen

Estrone
tro

Figure 1. Normal Endometrium and Endometriosis.


In normal endometrial tissue (Panel A), activity of the enzyme cyclooxygenase-2 (COX-2), and thus production
of prosCOLOR FIGURE
taglandin E2 (PGE2), is low. Estrogen is not produced locally, owing to the absence of aromatase.
the12/29/08
luteal
DraftDuring
5
phase, the progesterone-dependent 17-hydroxysteroid dehydrogenase 2 (HSD17B2) enzymeAuthor
catalyzes
the conversion
Bulun
1 endometriosis
Fig #
of the biologically active estradiol to estrone that is less estrogenic. In the endometrium of women
with
Endometriosis
Title
(Panel B), there is a subtle increase in COX-2 activity and detectable aromatase activity. In ectopic
endometriotic
tissue
ME
(Panel C), full-blown molecular abnormalities include high COX-2 and aromatase levels. Increased PGE2 formation in
DE
Schwartz
endometrial and endometriotic tissues can cause severe menstrual cramps and chronic pelvic
Tissue estradiol
Artist pain.Knoper
AUTHOR to
PLEASE
NOTE:
levels should be high, because estradiol is overproduced by aromatase and is not metabolized owing
deficient
Figure has been redrawn and type has been reset
PleaseBulun
check carefully
HSD17B2 activity. Increasing enzyme activity is denoted by increasing thickness of arrows. Modified from
et al.25
Issue date

T-cell expressed and secreted) attract the granulocytes, natural killer cells, and macrophages that
are typical of endometriosis.1,32-36,44,45 Autoregulatory positive-feedback loops ensure further accumulation of these immune cells, cytokines, and
chemokines in established lesions.46
In patients with endometriosis, inflammatory
and immune responses, angiogenesis, and apoptosis are altered in favor of the survival and replenishment of endometriotic tissue.38,39,47-49 These
basic pathologic processes depend in part on estrogen or progesterone. Excessive formation of
estrogen and prostaglandin and the development
of progesterone resistance have emerged as clinically useful points of study because the therapeutic targeting of aromatase in the estrogen biosynthetic pathway, cyclooxygenase-2 (COX-2) in the
270

1/15/09

prostaglandin pathway, or the progesterone receptor reduces pelvic pain or laparoscopically visible
endometriosis or both (Fig. 1).9,20,50,51 These three
critical targets have been linked through specific
epigenetic markers (hypomethylation) that cause
overexpression of the nuclear receptors steroidogenic factor 1 (SF1) and estrogen receptor .24,52
Estrogen Formation in Endometriosis

Estrogen production plays a key role in endometriosis. Its inhibition by GnRH analogues, oral contraceptives, progestins, and aromatase inhibitors
reduces pelvic disease and pain (Fig. 2A).16 Steroidogenic acute regulatory protein (STAR) facilitates the initial step of estrogen formation (i.e., the
entry of cytosolic cholesterol into the mitochondrion). Then, five proteins catalyzing six enzymatic

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

Mechanisms of Disease

A Sources of estradiol in endometriotic tissue


Pituitary
FSH
LH

GnRH analogues
Oral contraceptives
Progestins

Endometriosis

Induction of
gene expression

Cholesterol

Cholesterol

Adrenal gland

Androstenedione

Androstenedione

Androstenedione

Aromatase

Aromatase
inhibitor

Aromatase

Aromatase

Estrone

Estrone

Estrone

1-Estradiol

3-Estradiol

2-Estradiol

Ovary

Fat and skin

B Survival and inflammation of endometriotic tissue


Interleukin-1

VEGF
+

+
Cholesterol
STAR

Inflammation

PGE2

COX-2

Arachidonic acid

+
SCC
+

SF1

ER-

Estradiol

HSD3B2
17-hydrolyase
17-20-lyase
Androstenedione

Survival

HSD17B1
Aromatase

Estrone

Figure 2. Molecular Distinctions between Endometriotic Tissue and Endometrium.


Panel A shows the three sources of estradiol, biologically active estrogen, in endometriotic tissue. The
COLORfirst
FIGURE sources
Draft 8 of ovarian
12/29/08
are follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which induce the expression
steAuthor
Bulun
roidogenic genes, including aromatase, for biosynthesis of estradiol. Ovarian secretion of estradiol
can be reduced
2
Fig #
through suppression of FSH and LH by gonadotropin-releasing hormone (GnRH) analogues,
combination
oral conEndometriosis
Title
traceptives, or progestins. The second source of estrogen is the estradiol that arises from aromatase
activity in fat
ME
or skin. The third source of estradiol is local production in endometriotic tissue. Aromatase
in peripheral
DEinhibition
Schwartz
Artistin endometriosis.
Knoper
tissue (fat and skin) and endometriotic tissue stops estradiol biosynthesis and is therapeutic
As
AUTHOR PLEASE NOTE:
shown in Panel B, high levels of local estradiol and prostaglandin E2 (PGE2) are maintainedFigure
in endometriotic
tissue
has been redrawn and type has been reset
Please check carefully
by autoregulatory positive-feedback mechanisms (indicated by plus signs) that involve nuclear receptors
(steroidoIssue date 1/15/09
genic factor 1 [SF1] and estrogen receptor [ER-]), enzymatic pathways, cytokines, and growth factors. COX-2 denotes cyclooxygenase-2, HSD17B1 17-hydroxysteroid dehydrogenase 1, HSD3B2 3-hydroxysteroid dehydrogenase
2, SCC side-chain cleavage enzyme, STAR steroidogenic acute regulatory protein, and VEGF vascular endothelial
growth factor. Modified from Bulun et al.25

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

271

The

n e w e ng l a n d j o u r na l

steps (side-chain cleavage enzyme, 3-hydroxy


steroid dehydrogenase 2, 17-hydroxylase17-20lyase, aromatase, and 17-hydroxysteroid dehydrogenase 1) convert cholesterol to biologically
active estradiol (Fig. 2B).25 The key step, the conversion of C19 steroids to estrogens, is catalyzed
by aromatase, inhibition of which effectively eliminates all estrogen production (Fig. 2B).25
Origins of Estrogen in Endometriosis

Three major sites in the body produce estrogen in


women with endometriosis. First, estradiol secreted by the ovary reaches endometriotic tissue
through the circulation. Moreover, follicular rupture during each ovulation causes spillage of large
amounts of estradiol directly onto pelvic implants.
Second, aromatase in adipose tissue and skin catalyzes the conversion of circulating androstenedione to estrone that is subsequently converted to
estradiol, and this estrone and estradiol can enter
the circulation and reach sites of endometriosis.
Another source of estradiol is cholesterol, which
is converted to estradiol locally in endometriosis
because endometriotic tissue expresses a complete
set of steroidogenic genes, including aromatase.25
Peripheral or local aromatase activity, or both, may
be particularly important in the persistence of endometriosis the use of a combination of a GnRH
agonist plus an aromatase inhibitor is superior to
the use of a GnRH agonist alone for long-lasting
relief of pain (Fig. 2A).53
Estrogen Production and Inflammation

Cell survival and inflammation are responsible


for chronic pelvic pain and infertility, the primary
symptoms of endometriosis. Estrogen enhances
the survival or persistence of endometriotic tissue,
whereas prostaglandins and cytokines mediate
pain, inflammation, and infertility.54,55 A link between inflammation and estrogen production in
endometriosis was uncovered in a feedback cycle
that favors the overexpression of key steroidogenic
genes (most notably aromatase), overexpression
of COX-2, and continuous local production of estradiol and prostaglandin E2 in endometriotic
tissue.41,56-58
Prostaglandin E2 and Biosynthesis of Estradiol

Prostaglandin E2 coordinately stimulates the expression of all steroidogenic genes necessary to enable the endometriotic stromal cell to synthesize
estradiol from cholesterol.25 Among the six ste272

of

m e dic i n e

roidogenic genes, the regulation of two STAR


and CYP19A1, the aromatase gene has been characterized extensively (Fig. 2B).25
Endometriotic and endometrial stromal cells
express each of the prostaglandin E2 receptor subtypes, EP1, EP2, EP3, and EP4.59 Activation of the
EP2 receptor raises the intracellular levels of cyclic
AMP (cAMP), which is responsible for inducing the
expression of STAR and CYP19A1 by way of prostaglandin E2 in endometriotic stromal cells.25,57,59
Prostaglandin E2 or a cAMP analogue increases
STAR and aromatase levels and activity in endometriotic cells but not in endometrial stromal
cells.25,41,57 Thus, steroidogenesis dependent on
prostaglandin E2 and cAMP in endometriotic stro
mal cells requires downstream effectors, whereas
inhibitory mechanisms or a lack of stimulatory
effectors downstream of cAMP account for the
absence of steroidogenesis in normal endometrial
stromal cells.
The nuclear receptor SF1, which is present in
endometriotic tissue and absent in endometrium,
is the key transcription factor that mediates the
expression dependent on prostaglandin E2 and
cAMP of STAR, CYP19A1, and possibly other
steroidogenic genes in endometriotic stromal cells.
In prostaglandin E2 treated endometriotic cells,
SF1 binds coordinately and assembles an enhancer
transcriptional complex recruited to the promoters
of the STAR and CYP19A1 genes to induce their
expression (Fig. 2B).25
The absence of SF1 in endometrial cells is a
major cause of the lack of responsiveness of steroidogenic genes to prostaglandin E2. In addition,
the redundancy of transcriptional inhibitors of
STAR and CYP19A1 gene promoters in endometrial
cells constitute a fail-safe system for silencing
these steroidogenic genes. These repressors are
chicken ovalbumin upstream promotertranscription factor (COUP-TF), the Wilms tumor 1 transcription factor (WT1), and CCAAT/enhancer binding protein (C/EBP). Levels of WT1 and C/EBP
are much higher in normal endometrium than in
endometriotic tissue. In the absence of SF1, a transcriptional complex composed of repressors binds
the steroidogenic promoters and suppresses them
in endometrial cells.25
In summary, on induction of prostaglandin E2,
coordinated recruitment of SF1 to the promoters
of the essential steroidogenic genes is the key event
for estradiol synthesis in endometriotic stromal
cells. A suitable therapeutic target among the ste-

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

Mechanisms of Disease

roidogenic enzymes is aromatase, because its inhibition blocks all estradiol biosynthesis. Aromatase
inhibitors diminish or eradicate endometriotic
implants and associated pain that is refractory to
currently available treatments.19
Prostaglandin Production in Endometriosis

Prostaglandins, locally produced hormones involved in inflammation and pain, are important
in the pathogenesis of endometriosis. In particular, prostaglandin E2 and prostaglandin F2 are
produced in excess in uterine and endometriotic
tissues of women with endometriosis.60 The vasoconstrictive properties of prostaglandin F2, together with its ability to cause uterine contractions,
contributes to dysmenorrhea, whereas prostaglandin E2 can induce pain directly.60 These prostaglandins are clinically relevant because the reduction of prostaglandin formation by nonselective
COX inhibitors (e.g., naproxen sodium and ibuprofen) decreases pelvic pain associated with endometriosis.16,50 Care must be taken in long-term
administration of nonselective COX inhibitors because they have gastrointestinal side effects. Selective COX-2 inhibitors with milder side effects
(e.g., rofecoxib and valdecoxib) were approved by
the Food and Drug Administration for the treatment of primary dysmenorrhea, and their longterm administration could potentially reduce the
chronic pelvic pain associated with endometriosis.
The use of COX-2 inhibitors has, however, been
limited by their link to an increased risk of cardiovascular disease.50
Cyclooxygenase catalyzes the conversion of
arachidonic acid (released by phospholipase A2)
to prostaglandin H2 in most cells, including those
in myometrium, endometrium, and endometriotic
tissue.61 Of the two isoforms, COX-1 is generally
responsible for basal prostaglandin synthesis,
whereas COX-2 is important in inflammation.
The coupling of COX-dependent synthesis of prostaglandin H2 to its metabolism by enzymes downstream is orchestrated in a cell-specific fashion.
Uterine cells are rich in prostaglandin F synthase and microsomal prostaglandin E synthase,
which catalyze the conversion of prostaglandin
H2 to prostaglandin F2 and prostaglandin E2,
respectively.60
Excessive prostaglandin E2 production during
inflammation through coordinated induction of
multiple enzymes, in particular COX-2 and microsomal prostaglandin E synthase, is a concept

in development.61,62 Endometriotic stromal cells


produce large quantities of prostaglandin E2,
which induce local estrogen biosynthesis and pelvic pain.41,56,57,63 COX-2 is up-regulated to a greater
degree in endometriotic stromal cells as compared
with endometrial stromal cells; moreover, its expression is also increased in the endometrium of
women with endometriosis as compared with that
of disease-free women.64,65 Expression of micro
somal prostaglandin E synthase has been shown
in both benign and malignant endometrium.66-68
Thus, increased synthesis of prostaglandin E2 in
endometriotic tissue may be due to coordinated
hyperactivity of COX-2 and microsomal prostaglandin E synthase.
At least four hormones, present in high quantities in endometriotic tissue, induce COX-2 expression and prostaglandin E2 production in endometriotic and uterine cells (Fig. 2B). The cytokine
interleukin-1 or prostaglandin E2 itself (in an
autocrine manner) induces COX-2 expression in
endometriotic and endometrial stromal cells.69,70
The angiogenic factor vascular endothelial growth
factor or estradiol rapidly induces, through estrogen receptor , COX-2 expression in uterine endothelial and endometriotic stromal cells.69-71
These redundant mechanisms maintain large
quantities of prostaglandin E2 in endometriotic
tissue (Fig. 2B).
Thus, cytokines, angiogenic factors, prostaglandin E2 itself, and estradiol all induce COX-2
expression in patients with endometriosis and
thereby ensure production of large quantities of
prostaglandin E2. Estrogen receptor mediates
the induction of COX-2 by estradiol in endometriotic tissue.
Progesterone Resistance in Endometriosis

In contrast to the clearly unfavorable effect of estrogen on endometriosis, the role of progesterone
has remained ambiguous for the following three
reasons. First, the protective role of progesterone
with regard to endometrial cancer, in which there
is epithelial-cell proliferation, has been inappropriately attributed to endometriosis, which consists
primarily of stromal cells with a low rate of apoptosis and little differentiation.49 Paradoxically, progesterone induces the transient proliferation of
stromal cells in normal endometrium during the
secretory phase. Second, progestin-based treatments for pain are at best variably effective, especially in patients who have previously received other

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

273

The

Estradiol

HSD17B2

RA
XR
R
RAR RXR
Sp1
DNA

Sp3

n e w e ng l a n d j o u r na l

Estrone
tro

HSD17B2
mRNA

X
HSD17B2
promoter

Epithelium

Stroma

Other
paracrine factors

CH3
C

RA

X
Lack of binding

X
PR

Progesterone
Figure 3. Disrupted Paracrine Action of Progesterone in Endometriotic
Tissue.
COLOR FIGURE
In endometriotic tissue, the paracrine action of progesterone
is disrupted
Draft 5
12/29/08
by the decreased progesterone receptor (PR) levels
in the Bulun
stromal cell. In
Author
3
contrast, in the endometrium, progesterone induces
of epiFig #the expression
Title
thelial 17-hydroxysteroid dehydrogenase 2 (HSD17B2)
byEndometriosis
activating stromal PRs. These receptors mediate the formation of ME
retinoic acid (RA) and
DE
Schwartz
other unknown paracrine factors, which induce the
binding
to, and thus acArtist
Knoper
tivation of, the HSD17B2 promoter by a transcriptional AUTHOR
activator
complex
of
PLEASE
NOTE:
Figure has been redrawn and type has been reset
the transcription factors Sp1 and Sp3, retinoic acid receptor
(RAR),
and
Please
check
carefully
retinoid X receptor (RXR). The outcome is a decrease
local biologicalIssue date in
1/15/09
ly active estrogen, estradiol. The term mRNA denotes messenger RNA.

forms of treatment.72,73 Progestins probably reduce


pain by suppressing or attenuating ovulation. A direct effect of progestins on endometriotic tissue
cannot be ruled out, however. Third, a spectrum
of antiprogestins with a mix of agonist and an274

of

m e dic i n e

tagonist properties (selective progesterone-receptor


modulators) may reduce endometriosis-associated
pelvic pain more effectively than progestins.74,75
To add a further twist, endometriotic tissue produces large quantities of progesterone and contains much lower levels of progesterone receptors
than endometrium.23,57 These seemingly disparate
observations make it difficult to form a unified
hypothesis regarding the role of progesterone in
endometriosis.
Estrogen and progesterone are essential and
sufficient to control all endometrial function by
regulating expression of hundreds to thousands
of genes during the menstrual cycle.76 Indeed, the
administration of estradiol and progesterone is
sufficient to prepare the endometrium for implantation in postmenopausal women undergoing donor embryo transfer.77 Progesterone induces the
differentiation of endometrial stromal cells (called
decidualization) and epithelial cells (resulting in
the secretory phenotype). Molecular markers of
progesterone action include increased production
of epithelial glycodelin (a glycoprotein produced
by the secretory endometrium during the luteal
phase) and stromal prolactin in endometrium.76,78,79
Progesterone, however, induces much lower levels
of prolactin expression in endometriotic cells than
in endometrial stromal cells, suggesting that progesterone resistance may be at play in endome
triosis.80
In the endometrium, progesterone exerts an
antiestrogenic effect, in part by inducing 17-
hydroxysteroid dehydrogenase 2 (HSD17B2), which
catalyzes the conversion of biologically potent estradiol to the much less estrogenic estrone.81-84
Progesterone acts by way of progesterone receptors
in endometrial stromal cells to increase formation
of retinoic acid, which in turn induces HSD17B2
expression in endometrial epithelial cells, in a
paracrine fashion.85-87 Endometriotic stromal cells,
however, fail to respond to progesterone and thus
do not produce retinoic acid (Fig. 3).88 In endometriotic tissue, this lack of retinoic acid leads
to the lack of epithelial HSD17B2 and the failure
to inactivate estradiol.88,89 In combination with
high estradiol production due to aberrant aromatase activity, this additional defect contributes
to the abnormally high levels of estradiol in endometriotic tissue.80
Gene-expression profiles of the endometrium
of women with and those without endometriosis
have shown that a number of genes targeted by

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

Mechanisms of Disease

progesterone are deregulated during implantation,


at which time the endometrium is exposed to the
highest levels of progesterone.35,42 For example,
the prototypic gene responsive to progesterone,
glycodelin, is down-regulated in the endometrium
of women with endometriosis as compared with
women without endometriosis.35 These findings
suggest that eutopic endometrium of women with
endometriosis also exhibits progesterone resistance.35,90
Progesterone resistance is explicable by the extremely low progesterone-receptor levels in endometriotic tissue.23 In endometrium, levels of the
progesterone receptor isoforms, PR-B and PR-A,
progressively increase during the proliferative
phase, peak immediately before ovulation, and
diminish after ovulation, suggesting that estradiol
23 In constimulates progesterone-receptor levels.
Endometriosis
trast, PR-B is undetectable, and PR-A is markedly
reduced, in endometriotic tissues. Moreover, deficiency of the cochaperone FK506-binding protein of 52 kD, a peptidyl prolyl isomerase necessary for progesterone-receptor function in the
endometrium of baboons with endometriosis, may
contribute to progesterone resistance.91
Epigenetic Changes in Endometriosis

Levels of the nuclear receptors SF1, estrogen receptors and , and progesterone receptors are
much different in endometriotic tissue than in
endometrium. The virtual absence of the orphan
nuclear receptor SF1 in endometrium and its presence at levels more than 12,000 times higher, in
endometriotic tissue, is in part determined by a
classic CpG (cytosinephosphateguanine) island
at its promoter that is heavily methylated in endometrial stromal cells and unmethylated in endometriotic stromal cells.52 In endometrial cells,
the silencer-type transcription factor methyl-CpGbinding domain protein 2 is recruited to the methylated SF1 promoter and prevents its interaction
with transcriptional activators (Fig. 4). In endometriotic cells, the transcription factor, upstream
stimulatory factor 2, binds to the unmethylated
SF1 promoter and activates it.92 Upstream stimulatory factor 2 levels are much higher in endometriotic tissue than in the endometrium. Thus, differential SF1 expression in endometriotic tissue as
compared with endometrium is primarily controlled by an epigenetic mechanism that permits
the binding of activator complexes, rather than inhibitor complexes, to its promoter.52,92

Endometriotic tissue

Endometrium

Coactivator

Corepressor

MeCP2
SF1 or ER-
promoters
Methylated
CpG

Coding
region

SF1 or ER-
promoters

Coding
region

Unmethylated
CpG

SF1 or ER-

Arachidonic acid
COX-2

ER-

ER-

PGE2

PR

SF1

RA

Cholesterol

Aromatase

Estradiol

HSD17B2

Estrone

Figure 4. Epigenetic Changes in Endometriotic Tissue.


COLOR FIGURE
Draft
7
12/29/08
The promoters of two nuclear receptors, steroidogenic
factor
1 (SF1)
and
Bulun
estrogen receptor (ER-), are heavily methylatedAuthor
and thus
silenced in en4
Fig #
dometrial stromal cells. A lack of promoter methylation
is associated with
Endometriosis
Title
promoter activation and pathologic overexpression
of these nuclear recepME
tors in endometriotic stromal cells. SF1 mediates DE
prostaglandin
Schwartz E 2 (PGE 2)
Knoper estrogen redependent induction of estradiol production. ER-Artist
suppresses
AUTHOR PLEASE NOTE:
ceptor (ER-) and progesterone receptors (PRs),Figure
leading
to progesterone
has been redrawn
and type has been reset
Please check carefully
resistance and deficient inactivation of estradiol. Consequently,
in endodate 1/15/09
metriotic tissue, estradiol and PGE2 are producedIssue
in large
quantities and
enhance cell survival and inflammation. The corepressor and coactivator
are hypothetical. COX-2 denotes cyclooxygenase-2, CpG a cytosinephosphateguanine sequence, HSD17B2 17-hydroxysteroid dehydrogenase 2,
MeCP2 methyl-CpG-binding domain protein 2, and RA retinoic acid.

Among estrogen receptors, estrogen receptor


levels in endometriotic tissue are 142 times the
levels in endometrium, whereas estrogen receptor levels are 9 times the levels in endometrium.24 Hypomethylation of a CpG island at the
promoter region of the estrogen receptor gene
causes high levels of expression of the gene in
endometriotic stromal cells, and hypermethylation
silences it in endometrial stromal cells (Fig. 4).
Estrogen receptor in endometriotic stromal cells
occupies the estrogen receptor promoter and
down-regulates its activity, thus favoring the suppression of estrogen receptor levels.24 The high
ratio of estrogen receptor levels and estrogen
receptor levels in endometriotic stromal cells in
turn leads to increased estrogen receptor bind-

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

275

The

n e w e ng l a n d j o u r na l

of

m e dic i n e

Cross section of a 5-week, 46,XX embryo

Location of
cross section
Yolk sac

Wolffian
duct

Origin of
mllerian duct
Aorta

Migrating
germ cells

Genital
ridge

Genetic or
environmental effects
Epigenetic aberrations in progenitor cells
Hypomethylation of SF1 and ER- promoters
Subtle differentiation defects of uterus,
ovaries, or pelvic peritoneum

Endometriotic phenotype

Gonadal
ridge

Coelomic
cavity

Hindgut

Figure 5. Speculated Changes in DNA Methylation from Genetic or Environmental Factors.


COLOR FIGURE
Draft factors
6
12/29/08
During the embryonic differentiation of the female genital tract, various environmental or genetic
may
cause
Author
Bulun
changes in DNA methylation, shown here for a 5-week embryo. If these epigenetic alterations
pathologically
affect
5
Fig #
the expression of critical genes such as steroidogenic factor 1 (SF1) or estrogen receptor Title
(ER-) in
progenitor
Endometriosis
cells destined to form various pelvic tissues, the ensuing molecular abnormalities may predispose
the
adult woman
ME
to endometriosis.
DE
Schwartz
Artist

Knoper
AUTHOR PLEASE NOTE:

Figure has been redrawn and type has been reset


Please check carefully

ing to the progesterone-receptor promoter and endometrium, whereas theIssue


lack
of methylation redate 1/15/09
mediates the down-regulation of expression of sults in extraordinarily high levels of SF1 and esprogesterone receptors (Fig. 4).
trogen receptor in endometriotic tissue (Fig. 4).
In response to the exposure of endometriotic cells
to prostaglandin E2, SF1 coordinately binds to the
C onclusions
promoters of multiple steroidogenic genes, includIncreased cell survival, inflammation, and defi- ing that of aromatase, and causes the formation of
cient differentiation in endometriosis have been large quantities of estradiol. Estradiol acts by way
linked to a stromal-cell defect involving the exces- of estrogen receptor to stimulate COX-2, leading
sive formation of estrogen and prostaglandin, as to the overproduction of prostaglandin E2. Thus,
well as progesterone resistance, all of which origi- inflammation and estrogen are linked in a feednate from two distinct epigenetic changes that af- back cycle involving the overexpression of genes
fect the transcription factors SF1 and estrogen re- that encode the aromatase and COX-2 enzymes
ceptor . The CpG islands occupying the promoters and continuous formation of the products of aroof the genes encoding SF1 and estrogen receptor matase and COX-2 estradiol and prostaglandin
are heavily methylated and silence these genes in E2 in endometriotic tissue. Moreover, estrogen
276

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

Mechanisms of Disease

receptor suppresses progesterone-receptor levels, resulting in progesterone resistance and disruption of a paracrine pathway that inactivates estradiol. Large amounts of estradiol accumulate,
owing to its increased formation and deficient
inactivation in endometriotic tissue (Fig. 4).
This working model (Fig. 4) is clinically relevant because the targeting of aromatase, COX-2,
estrogen receptor , or progesterone receptors reduces pelvic pain and ablates visible endometriotic
tissue.93 Finally, I speculate that genetic predisposition or the exposure to environmental toxins
References
1. Giudice LC, Kao LC. Endometriosis.
Lancet 2004;364:1789-99.
2. Rock JA, Zacur HA, Dlugi AM, Jones
HW Jr, TeLinde RW. Pregnancy success
following surgical correction of imperforate hymen and complete transverse vaginal septum. Obstet Gynecol 1982;59:44851.
3. Missmer SA, Hankinson SE, Spiegelman
D, Barbieri RL, Michels KB, Hunter DJ. In
utero exposures and the incidence of endometriosis. Fertil Steril 2004;82:1501-8.
4. Simpson JL, Elias S, Malinak LR, Buttram VC Jr. Heritable aspects of endometriosis. I. Genetic studies. Am J Obstet
Gynecol 1980;137:327-31.
5. Montgomery GW, Nyholt DR, Zhao
ZZ, et al. The search for genes contributing to endometriosis risk. Hum Reprod
Update 2008;14:447-57.
6. Treloar SA, Wicks J, Nyholt DR, et al.
Genomewide linkage study in 1,176 affected sister pair families identifies a significant susceptibility locus for endometriosis on chromosome 10q26. Am J
Hum Genet 2005;77:365-76.
7. Garry R. The endometriosis syndromes:
a clinical classification in the presence of
aetiological confusion and therapeutic anarchy. Hum Reprod 2004;19:760-8.
8. Brosens I. Endometriosis rediscovered? Hum Reprod 2004;19:1679-80.
9. Guzick DS, Silliman NP, Adamson
GD, et al. Prediction of pregnancy in infertile women based on the American Society for Reproductive Medicines revised
classification of endometriosis. Fertil Steril 1997;67:822-9.
10. Stovall DW, Bowser LM, Archer DF,
Guzick DS. Endometriosis-associated pelvic pain: evidence for an association between the stage of disease and a history of
chronic pelvic pain. Fertil Steril 1997;68:
13-8. [Erratum, Fertil Steril 1998;69:979.]
11. Berkley KJ, Dmitrieva N, Curtis KS,
Papka RE. Innervation of ectopic endometrium in a rat model of endometriosis. Proc
Natl Acad Sci U S A 2004;101:11094-8.
12. Barnhart K, Dunsmoor-Su R, Coutifaris C. Effect of endometriosis on in vitro
fertilization. Fertil Steril 2002;77:1148-55.

of fetal progenitor cells destined to form adult


female pelvic organs may result in epigenetic
events, including promoter hypomethylation and
overexpression of SF1 and estrogen receptor ,
that can play critical roles in the pathogenesis of
endometriosis (Fig. 5).
Supported in part by grants from the National Institutes of
Health (HD38691 and HD40093) and from the Friends of Prentice.
Dr. Bulun reports receiving consultation fees from Meditrina
Pharmaceuticals, GlaxoSmithKline, and Novartis. No other potential conflict of interest relevant to this article was reported.
I thank Dr. Sherman Elias for helpful discussions.

13. Somigliana E, Vigan P, Parazzini F,

Stoppelli S, Giambattista E, Vercellini P.


Association between endometriosis and
cancer: a comprehensive review and a critical analysis of clinical and epidemiological evidence. Gynecol Oncol 2006;101:
331-41.
14. Dinulescu DM, Ince TA, Quade BJ,
Shafer SA, Crowley D, Jacks T. Role of
K-ras and Pten in the development of
mouse models of endometriosis and endometrioid ovarian cancer. Nat Med 2005;
11:63-70.
15. Olive DL, Schwartz LB. Endometriosis. N Engl J Med 1993;328:1759-69.
16. Olive DL, Pritts EA. Treatment of endometriosis. N Engl J Med 2001;345:26675.
17. Missmer SA, Hankinson SE, Spiegelman
D, et al. Reproductive history and endometriosis among premenopausal women.
Obstet Gynecol 2004;104:965-74.
18. Takayama K, Zeitoun K, Gunby RT,
Sasano H, Carr BR, Bulun SE. Treatment
of severe postmenopausal endometriosis
with an aromatase inhibitor. Fertil Steril
1998;69:709-13.
19. Attar E, Bulun SE. Aromatase inhibitors: the next generation of therapeutics
for endometriosis? Fertil Steril 2006;85:
1307-18.
20. Kettel LM, Murphy AA, Morales AJ,
Ulmann A, Baulieu EE, Yen SS. Treatment
of endometriosis with the antiprogesterone mifepristone (RU486). Fertil Steril
1996;65:23-8.
21. Murphy AA, Castellano PZ. RU486:
pharmacology and potential use in the
treatment of endometriosis and leiomyomata uteri. Curr Opin Obstet Gynecol
1994;6:269-78.
22. Brandenberger AW, Lebovic DI, Tee
MK, et al. Oestrogen receptor (ER)-alpha
and ER-beta isoforms in normal endometrial and endometriosis-derived stromal
cells. Mol Hum Reprod 1999;5:651-5.
23. Attia GR, Zeitoun K, Edwards D,
Johns A, Carr BR, Bulun SE. Progesterone
receptor isoform A but not B is expressed
in endometriosis. J Clin Endocrinol Metab
2000;85:2897-902.

24. Xue Q, Lin Z, Cheng YH, et al. Pro-

moter methylation regulates estrogen receptor 2 in human endometrium and endometriosis. Biol Reprod 2007;77:681-7.
25. Bulun SE, Lin Z, Imir G, et al. Regulation of aromatase expression in estrogenresponsive breast and uterine disease:
from bench to treatment. Pharmacol Rev
2005;57:359-83.
26. Sampson JA. Peritoneal endometriosis
due to menstrual dissemination of endometrial tissue into the peritoneal cavity.
Am J Obstet Gynecol 1927;14:422-69.
27. DHooghe TM, Debrock S. Endometriosis, retrograde menstruation and peritoneal inflammation in women and in baboons. Hum Reprod Update 2002;8:84-8.
28. Ferguson BR, Bennington JL, Haber
SL. Histochemistry of mucosubstances
and histology of mixed mllerian pelvic
lymph node glandular inclusions: evidence
for histogenesis by mllerian metaplasia
of coelomic epithelium. Obstet Gynecol
1969;33:617-25.
29. Sampson JA. Metastatic or embolic
endometriosis due to menstrual dissemination of endometrial tissue into the venous circulation. Am J Pathol 1927;3:93109.
30. Sasson IE, Taylor HS. Stem cells and
the pathogenesis of endometriosis. Ann
N Y Acad Sci 2008;1127:106-15.
31. Lucidi RS, Witz CA, Chrisco M, Binkley PA, Shain SA, Schenken RS. A novel
in vitro model of the early endometriotic
lesion demonstrates that attachment of
endometrial cells to mesothelial cells is
dependent on the source of endometrial
cells. Fertil Steril 2005;84:16-21.
32. Tseng JF, Ryan IP, Milam TD, et al.
Interleukin-6 secretion in vitro is up-regulated in ectopic and eutopic endometrial
stromal cells from women with endometriosis. J Clin Endocrinol Metab 1996;
81:1118-22.
33. Hornung D, Ryan IP, Chao VA, Vigne
JL, Schriock ED, Taylor RN. Immunolocalization and regulation of the chemokine
RANTES in human endometrial and endometriosis tissues and cells. J Clin Endocrinol Metab 1997;82:1621-8.

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

277

The

n e w e ng l a n d j o u r na l

34. Wu Y, Kajdacsy-Balla A, Strawn E, et

al. Transcriptional characterizations of


differences between eutopic and ectopic
endometrium. Endocrinology 2006;147:
232-46.
35. Kao LC, Germeyer A, Tulac S, et al.
Expression profiling of endometrium from
women with endometriosis reveals candidate genes for disease-based implantation
failure and infertility. Endocrinology 2003;
144:2870-81.
36. Osteen KG, Bruner KL, Sharpe-Timms
KL. Steroid and growth factor regulation
of matrix metalloproteinase expression
and endometriosis. Semin Reprod Endocrinol 1996;14:247-55.
37. Nair A, Nair H, Lucidi R, et al. Modeling the early endometriotic lesion: mesothelium-endometrial cell co-culture increases endometrial invasion and alters
mesothelial and endometrial gene transcription. Fertil Steril 2007;90:1487-95.
38. Dmowski WP, Gebel HM, Rawlins
RG. Immunologic aspects of endometriosis. Obstet Gynecol Clin North Am 1989;
16:93-103.
39. Osteen KG, Sierra-Rivera E. Does disruption of immune and endocrine systems by environmental toxins contribute
to development of endometriosis? Semin
Reprod Endocrinol 1997;15:301-8.
40. Noble LS, Simpson ER, Johns A, Bulun SE. Aromatase expression in endometriosis. J Clin Endocrinol Metab 1996;
81:174-9.
41. Noble LS, Takayama K, Zeitoun KM,
et al. Prostaglandin E2 stimulates aromatase expression in endometriosis-derived
stromal cells. J Clin Endocrinol Metab
1997;82:600-6.
42. Burney RO, Talbi S, Hamilton AE, et
al. Gene expression analysis of endometrium reveals progesterone resistance and
candidate susceptibility genes in women
with endometriosis. Endocrinology 2007;
148:3814-26.
43. Sharpe-Timms KL, Cox KE. Paracrine
regulation of matrix metalloproteinase expression in endometriosis. Ann N Y Acad
Sci 2002;955:147-56.
44. Ulukus M, Ulukus EC, Seval Y, Zheng
W, Arici A. Expression of interleukin-8
receptors in endometriosis. Hum Reprod
2005;20:794-801.
45. Akoum A, Jolicoeur C, Boucher A. Estradiol amplifies interleukin-1-induced
monocyte chemotactic protein-1 expression by ectopic endometrial cells of women with endometriosis. J Clin Endocrinol
Metab 2000;85:896-904.
46. Lebovic DI, Mueller MD, Taylor RN.
Immunobiology of endometriosis. Fertil
Steril 2001;75:1-10.
47. Taylor RN, Lebovic DI, Mueller MD.
Angiogenic factors in endometriosis. Ann
N Y Acad Sci 2002;955:89-100.
48. Dmowski WP, Ding J, Shen J, Rana N,
Fernandez BB, Braun DP. Apoptosis in endometrial glandular and stromal cells in

278

of

m e dic i n e

women with and without endometriosis.


Hum Reprod 2001;16:1802-8.
49. Bliard A, Nol A, Foidart JM. Reduction of apoptosis and proliferation in endometriosis. Fertil Steril 2004;82:80-5.
50. Hayes EC, Rock JA. COX-2 inhibitors
and their role in gynecology. Obstet Gynecol Surv 2002;57:768-80.
51. Vercellini P, Trespidi L, Colombo A,
Vendola N, Marchini M, Crosignani P.
A gonadotropin-releasing hormone agonist versus a low-dose oral contraceptive
for pelvic pain associated with endometriosis. Fertil Steril 1993;60:75-9.
52. Xue Q, Lin Z, Yin P, et al. Transcriptional activation of steroidogenic factor-1
by hypomethylation of the 5 CpG island
in endometriosis. J Clin Endocrinol Metab
2007;92:3261-7.
53. Soysal S, Soysal M, Ozer S, Gul N,
Gezgin T. The effects of post-surgical administration of goserelin plus anastrozole
compared to goserelin alone in patients
with severe endometriosis: a prospective
randomized trial. Hum Reprod 2004;19:
160-7.
54. Ryan IP, Taylor RN. Endometriosis
and infertility: new concepts. Obstet Gynecol Surv 1997;52:365-71.
55. Bruner KL, Matrisian LM, Rodgers
WH, Gorstein F, Osteen KG. Suppression
of matrix metalloproteinases inhibits establishment of ectopic lesions by human
endometrium in nude mice. J Clin Invest
1997;99:2851-7.
56. Tsai SJ, Wu MH, Lin CC, Sun HS,
Chen HM. Regulation of steroidogenic
acute regulatory protein expression and
progesterone production in endometriotic
stromal cells. J Clin Endocrinol Metab
2001;86:5765-73.
57. Sun HS, Hsiao KY, Hsu CC, Wu MH,
Tsai SJ. Transactivation of steroidogenic
acute regulatory protein in human endometriotic stromal cells is mediated by the
prostaglandin EP2 receptor. Endocrinology 2003;144:3934-42.
58. Bulun SE, Yang S, Fang Z, et al. Role
of aromatase in endometrial disease. J Steroid Biochem Mol Biol 2001;79:19-25.
59. Zeitoun KM, Bulun SE. Aromatase: a
key molecule in the pathophysiology of
endometriosis and a therapeutic target.
Fertil Steril 1999;72:961-9.
60. Jabbour HN, Sales KJ, Smith OP, Battersby S, Boddy SC. Prostaglandin receptors are mediators of vascular function in
endometrial pathologies. Mol Cell Endocrinol 2006;252:191-200.
61. Funk CD. Prostaglandins and leuko
trienes: advances in eicosanoid biology.
Science 2001;294:1871-5.
62. Mancini JA, Blood K, Guay J, et al.
Cloning, expression, and up-regulation of
inducible rat prostaglandin E synthase
during lipopolysaccharide-induced pyresis and adjuvant-induced arthritis. J Biol
Chem 2001;276:4469-75.
63. Zeitoun K, Takayama K, Michael MD,

Bulun SE. Stimulation of aromatase P450


promoter (II) activity in endometriosis and
its inhibition in endometrium are regulated by competitive binding of steroidogenic factor-1 and chicken ovalbumin upstream
promoter transcription factor to the same
cis-acting element. Mol Endocrinol 1999;
13:239-53.
64. Wu MH, Wang CA, Lin CC, Chen LC,
Chang WC, Tsai SJ. Distinct regulation of
cyclooxygenase-2 by interleukin-1beta in
normal and endometriotic stromal cells.
J Clin Endocrinol Metab 2005;90:286-95.
65. Ota H, Igarashi S, Sasaki M, Tanaka
T. Distribution of cyclooxygenase-2 in eutopic and ectopic endometrium in endometriosis and adenomyosis. Hum Reprod
2001;16:561-6.
66. Ni H, Sun T, Ding NZ, Ma XH, Yang
ZM. Differential expression of microsomal prostaglandin E synthase at implantation sites and in decidual cells of mouse
uterus. Biol Reprod 2002;67:351-8.
67. Sun T, Li SJ, Diao HL, Teng CB, Wang
HB, Yang ZM. Cyclooxygenases and prostaglandin E synthases in the endometrium
of the rhesus monkey during the menstrual cycle. Reproduction 2004;127:465-73.
68. Jabbour HN, Milne SA, Williams AR,
Anderson RA, Boddy SC. Expression of
COX-2 and PGE synthase and synthesis of
PGE(2) endometrial adenocarcinoma:
a possible autocrine/paracrine regulation
of neoplastic cell function via EP2/EP4 receptors. Br J Cancer 2001;85:1023-31.
69. Tamura M, Sebastian S, Yang S, Gurates B, Fang Z, Bulun SE. Interleukin1beta elevates cyclooxygenase-2 protein
level and enzyme activity via increasing
its mRNA stability in human endometrial
stromal cells: an effect mediated by extracellularly regulated kinase 1 and 2. J Clin
Endocrinol Metab 2002;87:3263-73.
70. Tamura M, Sebastian S, Yang S, et al.
Up-regulation of cyclooxygenase-2 expression and prostaglandin synthesis in
endometrial stromal cells by malignant
endometrial epithelial cells: a paracrine
effect mediated by prostaglandin E2 and
nuclear factor-kappa B. J Biol Chem 2002;
277:26208-16.
71. Tamura M, Sebastian S, Gurates B,
Yang S, Fang Z, Bulun SE. Vascular endothelial growth factor up-regulates cyclooxygenase-2 expression in human endothelial cells. J Clin Endocrinol Metab
2002;87:3504-7.
72. Vercellini P, Cortesi I, Crosignani PG.
Progestins for symptomatic endometriosis: a critical analysis of the evidence. Fertil Steril 1997;68:393-401.
73. Surrey ES. The role of progestins in
treating the pain of endometriosis. J Minim Invasive Gynecol 2006;13:528-34.
74. Chwalisz K, Perez MC, Demanno D,
Winkel C, Schubert G, Elger W. Selective
progesterone receptor modulator development and use in the treatment of leiomyomata and endometriosis. Endocr Rev

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

Mechanisms of Disease
2005;26:423-38. [Erratum, Endocr Rev
2005;26:703.]
75. Madauss KP, Grygielko ET, Deng SJ,
et al. A structural and in vitro characterization of asoprisnil: a selective progesterone receptor modulator. Mol Endocrinol
2007;21:1066-81.
76. Kao LC, Tulac S, Lobo S, et al. Global
gene profiling in human endometrium
during the window of implantation. Endocrinology 2002;143:2119-38.
77. Bulun SE, Adashi EY. The physiology
and pathology of the female reproductive
axis. In: Larsen PR, Kronenberg HM,
Melmed S, Polonsky K, eds. Williams
textbook of endocrinology. 10th ed. Philadelphia: W.B. Saunders, 2003:587-664.
78. Brosens JJ, Hayashi N, White JO. Progesterone receptor regulates decidual prolactin expression in differentiating human
endometrial stromal cells. Endocrinology
1999;140:4809-20.
79. Fazleabas AT, Brudney A, Chai D,
Langoi D, Bulun SE. Steroid receptor and
aromatase expression in baboon endometriotic lesions. Fertil Steril 2003;80:
Suppl 2:820-7.
80. Bulun SE, Cheng YH, Yin P, et al. Progesterone resistance in endometriosis:
link to failure to metabolize estradiol.
Mol Cell Endocrinol 2006;248:94-103.
81. Tseng L, Gurpide E. Estradiol and
20alpha-dihydroprogesterone dehydrogenase activities in human endometrium
during the menstrual cycle. Endocrinology 1974;94:419-23.
82. Tseng L, Gurpide E. Induction of human endometrial estradiol dehydroge-

nase by progestins. Endocrinology 1975;


97:825-33.
83. Satyaswaroop PG, Wartell DJ, Mortel
R. Distribution of progesterone receptor,
estradiol dehydrogenase, and 20 alphadihydroprogesterone dehydrogenase activities in human endometrial glands and
stroma: progestin induction of steroid dehydrogenase activities in vitro is restricted
to the glandular epithelium. Endocrinology 1982;111:743-9.
84. Yang S, Fang Z, Gurates B, et al.
Stromal PRs mediate induction of 17betahydroxysteroid dehydrogenase type 2 expression in human endometrial epithelium:
a paracrine mechanism for inactivation of
E2. Mol Endocrinol 2001;15:2093-105.
85. Casey ML, MacDonald PC, Andersson
S. 17 beta-Hydroxysteroid dehydrogenase
type 2: chromosomal assignment and progestin regulation of gene expression in
human endometrium. J Clin Invest 1994;
94:2135-41.
86. Mustonen MV, Isomaa VV, Vaskivuo T,
et al. Human 17beta-hydroxysteroid dehydrogenase type 2 messenger ribonucleic
acid expression and localization in term
placenta and in endometrium during the
menstrual cycle. J Clin Endocrinol Metab
1998;83:1319-24.
87. Cheng YH, Yin P, Xue Q, Yilmaz B,
Dawson MI, Bulun SE. Retinoic acid (RA)
regulates 17beta-hydroxysteroid dehydrogenase type 2 expression in endometrium:
interaction of RA receptors with specificity protein (SP) 1/SP3 for estradiol metabolism. J Clin Endocrinol Metab 2008;93:
1915-23.

88. Cheng YH, Imir A, Fenkci V, Yilmaz

MB, Bulun SE. Stromal cells of endometriosis fail to produce paracrine factors
that induce epithelial 17beta-hydroxysteroid dehydrogenase type 2 gene and its
transcriptional regulator Sp1: a mechanism for defective estradiol metabolism.
Am J Obstet Gynecol 2007;196:391.
89. Zeitoun K, Takayama K, Sasano H, et
al. Deficient 17beta-hydroxysteroid dehydrogenase type 2 expression in endometriosis: failure to metabolize 17betaestradiol. J Clin Endocrinol Metab 1998;
83:4474-80.
90. Osteen KG, Bruner-Tran KL, Eisenberg E. Reduced progesterone action during endometrial maturation: a potential
risk factor for the development of endometriosis. Fertil Steril 2005;83:529-37.
91. Jackson KS, Brudney A, Hastings JM,
Mavrogianis PA, Kim JJ, Fazleabas AT. The
altered distribution of the steroid hormone
receptors and the chaperone immunophilin FKBP52 in a baboon model of endometriosis is associated with progesterone
resistance during the window of uterine
receptivity. Reprod Sci 2007;14:137-50.
92. Utsunomiya H, Cheng YH, Lin Z, et al.
Upstream stimulatory factor-2 regulates
steroidogenic factor-1 expression in endometriosis. Mol Endocrinol 2008;22:904-14.
93. Harris HA, Bruner-Tran KL, Zhang X,
Osteen KG, Lyttle CR. A selective estrogen
receptor-beta agonist causes lesion regression in an experimentally induced model
of endometriosis. Hum Reprod 2005;20:
936-41.
Copyright 2009 Massachusetts Medical Society.

full text of all journal articles on the world wide web

Access to the complete text of the Journal on the Internet is free to all subscribers. To use this Web site, subscribers should go
to the Journals home page (NEJM.org) and register by entering their names and subscriber numbers as they appear on their
mailing labels. After this one-time registration, subscribers can use their passwords to log on for electronic access to the entire
Journal from any computer that is connected to the Internet. Features include a library of all issues since January 1993 and
abstracts since January 1975, a full-text search capacity, and a personal archive for saving articles and search results of interest.
All articles can be printed in a format that is virtually identical to that of the typeset pages. Beginning 6 months after
publication, the full text of all Original Articles and Special Articles is available free to nonsubscribers.

n engl j med 360;3 nejm.org january 15, 2009

The New England Journal of Medicine


Downloaded from nejm.org on June 3, 2015. For personal use only. No other uses without permission.
Copyright 2009 Massachusetts Medical Society. All rights reserved.

279

Вам также может понравиться