SlideShare a Scribd company logo
1 of 9
Download to read offline
© The Author 2006. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology. All rights reserved. For
Permissions, please email: journals.permissions@oxfordjournals.org 641
Human Reproduction Update, Vol.12, No.5 pp. 641–649, 2006 doi:10.1093/humupd/dml026
Advance Access publication June 14, 2006
Animal models in endometriosis research
Ruth Grümmer
Institute of Anatomy, University Hospital Essen, Essen, Germany
E-mail: ruth.gruemmer@uni-essen.de
Endometriosis is a common gynaecological disease, defined as the presence of endometrial tissue outside the uterus,
causing pelvic pain and subfertility in ~10% of women of reproductive age. Current therapies lead to pain relief,
however, do not address the causes and entail severe side effects. Still little is known about the pathogenic processes
leading to the development and maintenance of endometriosis. Because endometriosis occurs spontaneously only in
humans and some non-human primates, animal models of induced endometriosis have been developed and are of
high value for the evaluation of pathophysiological mechanisms underlying the development of this disease. These
experimental models include the autotransplantation of uterine fragments into the peritoneal cavity of rodents and
non-human primates or the heterotransplantation of human endometrial or endometriotic tissue to immunodeficient
mice or onto the chicken chorioallantoic membrane (CAM). This review describes the animal models for endometrio-
sis and assesses their different potentials and limitations in regard to endometriosis research, with the aim of develop-
ing novel non-invasive diagnostic tools and improved strategies for the treatment of endometriosis in women.
Key words: animal model/CAM/endometriosis/non-human primates/rodents
The need for animal models in endometriosis research
Endometriosis is one of the most frequent benign gynaecological
diseases; it is thought to occur in 7–10% of women (Wheeler,
1989) but may even affect up to 60% of women of reproductive
age with pelvic symptoms or disturbance of fertility (Pellicer
et al., 2001; Giudice and Kao, 2004). Yet, little is known about the
pathogenic processes leading to the development and maintenance
of this disease, and currently available medical therapies are unsat-
isfactory, because they focus on treating the symptoms rather than
curing the causes. In addition, they cannot be used for prolonged
duration because of severe secondary side effects (Rice, 2002).
Thus, there is a definite need to develop new drugs to provide spe-
cific and more efficient therapeutic alternatives that eliminate
endometriotic lesions, prevent recurrences and do not interfere
with the fertility potential.
One widely accepted mechanism for the development of perito-
neal endometriotic lesions is the adhesion and growth of endome-
trial fragments deposited into the peritoneal cavity via retrograde
menstruation (Sampson, 1927). However, because retrograde
menstruation occurs in nearly all women of reproductive age
(Halme et al., 1984), impairment of additional factors is needed
for the establishment and maintenance of these ectopic lesions.
Because, at the time of clinical presentation, most women already
have established endometriosis, it is hardly possible to give exper-
imental evidence for physiological roles in the pathogenesis of this
disease in humans. In addition, ethical considerations limit the
performance of controlled experiments, and it is not possible to
monitor the disease progression without performing repeated
laparoscopies. Thus, research into the fundamental mechanisms
by which menstrual endometrium adheres, invades and establishes
a functional vasculature to persist in an ectopic site, as well as the
development of new therapeutical approaches, is best performed
in experimental animal models.
Because menstrual shedding is a requirement for the spontane-
ous development of this disease, endometriosis occurs naturally
only in humans and some non-human primates. Non-human pri-
mates have been extensively used for the investigation of endome-
triosis; however, the very high costs of animal handling limit the
use of monkeys as an experimental model. For this reason, the
establishment of small laboratory animals, especially rodents, as
models for endometriosis by the transplantation of pieces of
endometrial tissue to ectopic sites has greatly extended in recent
years. This review provides a survey of commonly used animal
models and their application for different challenges.
CAM assay
Characterization of the CAM assay
The chicken chorioallantoic membrane (CAM) model can be con-
sidered as an animal model in the broader sense. Originally, it had
been developed to study the invasive, metastatic and angiogenic
potential of neoplastic cells (Scher et al., 1976; Armstrong et al.,
1982). It has been established as a model for endometriosis by cul-
turing fragments of human endometrial tissue on the basal layer of
the CAM of fertilized chicken eggs after prior incubation for 7–10
days (Malik et al., 2000; Maas et al., 2001). Endometrial fragments
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022
R.Grümmer
642
from the proliferative and secretory phase of the menstrual cycle
as well as the menstrual endometrium invade across the epithe-
lium into the mesenchymal layer and develop endometriosis-like
lesions in this layer of the CAM within 3 days after grafting of the
human tissue (Maas et al., 2001; Nap et al., 2003, 2004). It was
shown that these endometrial fragments needed to contain intact
glandular structures as well as stromal components.
Application of the CAM assay
Angiogenesis could be documented by ingrowth of vessels from
the CAM into the human endometrial fragments (Maas et al.,
2001). The anti-angiogenic factors endostatin, TNP-470 and
anti-vascular endothelial growth factor (VEGF) antibodies all
significantly decreased the angiogenic response to endometrial
transplants (Nap et al., 2005). Endometrial fragments of
women with and without endometriosis showed no significant
differences in VEGF expression or angiogenic induction in this
assay (Gescher et al., 2005). In contrast, Oosterlynck et al.
(1993) demonstrated that peritoneal fluid of women with
endometriosis induced significantly higher angiogenic
responses in the CAM than peritoneal fluid of women without
endometriosis.
Because the CAM contains extracellular matrix components
similar to the ECM of the human peritoneum (Giannopoulou
et al., 2001), it can be used to investigate the invasive potential
of endometriotic tissue. Nap et al. (2004) demonstrated that the
expression profiles of matrix metalloproteinases (MMPs) were
similar in experimentally induced endometriosis in CAMs
compared with human endometriosis and that the development
of early endometriotic lesions in the CAM model was pre-
vented by inhibiting MMP-1, -2, -3, -7 and -13 activity.
According to Wolber et al. (2003), relative concentrations of
MMP-1 mRNA, but not MMP-2 mRNA, increased strongly
after culture on the CAM.
Evaluation of the CAM assay
The CAM assay is well suited for the investigation of mechanisms
involved in invasion and angiogenesis during the first steps in the
establishment of endometriotic lesions, for example, by the inhibi-
tion of factors known to participate in these processes. The ectopic
lesions are easily accessible and can be simply monitored during
the course of an experiment. This endometriosis model is hardly
applicable for the investigation of immunological or inflammatory
responses or to analyse long-term drug effects in the course of the
disease.
Rodent models
In contrast to humans and non-human primates, estrous animals do
not shed their endometrial tissue and therefore do not develop
endometriosis spontaneously. However, endometriosis can be
induced by transplanting endometrial tissue to ectopic sites. These
models are classified into two types, homologous and heterologous
models. Homologous models have been employed utilizing the sur-
gical transplantation of endometrium of the same or syngeneic ani-
mals in immunocompetent animals, whereas in heterologous
models, human endometrial fragments are transferred either intra-
peritoneally or subcutaneously to immunodeficient mice.
Autotransplantation of endometrium to immunocompetent mice
and rats
Characterization of the homologous model
Autotransplantation of uterine tissue to ectopic sites of small labo-
ratory animals has been established not only in rats (Golan et al.,
1984; Jones, 1984; Vernon and Wilson, 1985; Rajkumar et al.,
1990; Sharpe et al., 1991) and mice (Cummings and Metcalf,
1995; Somigliana et al., 1999; Rossi et al., 2000) but also in ham-
sters (Steinleitner et al., 1991b) and rabbits (Schenken and Asch,
1980; Dunselman et al., 1989; Homm et al., 1989; Rock et al.,
1993). Of those non-primate models, mainly rat and mouse mod-
els have been further developed in recent years. In these models of
endometriosis, uteri are removed and cut into small pieces which
are then reintroduced, mostly by suturing, into the peritoneal cav-
ity. In most of these studies, endometrium was not separated from
myometrium; thus, both compartments were implanted (Vernon
and Wilson, 1985; Cummings and Metcalf, 1995). In the rat, the
uterine tissue develops into fluid-filled, ovoid, cystic structures
composed of myometrial and endometrial tissue. The cysts grow
but stabilize their size by ∼2 months and remain viable for at least
10 months (Vernon and Wilson, 1985).
In the mouse, the ectopic uterine fragments show histological
characteristics of the human disease, including the formation of
multiple, highly vascularized lesions containing endometrial
glands, stroma and cysts, independent of their peritoneal localiza-
tion in the abdomen (Cummings and Metcalf, 1995). A limited
number of experiments separating the endometrium from the myo-
metrium, only inoculating the endometrium to ectopic sites, have
been performed in rats (Katsuki et al., 1998) and mice
(Somigliana et al., 1999; Hirata et al., 2005; Yao et al., 2005). In
the study of Somigliana et al. (1999), isolated endometrial tissue
was minced and reintroduced into the peritoneal cavity of recipi-
ent, syngeneic animals. Both donor and recipient mice had been
ovariectomized and received estrogen treatment. In this study, all
recipient animals revealed evidence of peritoneal endometriosis
after 3 weeks, and neovascularization was observed at the surface
of the lesions. However, the ‘take-rate’, that is the number of
lesions recovered after a known number of endometrial fragments
have been randomly inoculated, was on average 30% of the inocu-
lated tissue.
For better identification of size and localization of ectopic
endometriotic lesions after transplantation, Hirata et al. (2005)
developed a homologous mouse model using ‘green mice’ that
ubiquitously express green fluorescent protein (GFP) as
donors and transplanted the fluorescent endometrial tissue into
recipient mice. They could show that the weight of endometriotic
lesions significantly correlated with the measured fluorescence
intensity. This fluorescence was significantly higher in the estrogen-
supplemented mice as compared with control animals, supporting
the estrogen dependency of these ectopic endometrial lesions.
Application of the homologous model
Endometriosis is known to be an estrogen-dependent disease in
women, and the suppression of the serum estrogen level supports
the regression of those ectopic lesions (Lapp, 2000; Rice, 2002).
Autotransplanted uterine tissue in the rodent model shows steroid
hormone dependency, and thus, this model has been widely used
for determining the responsiveness of ectopic lesions to steroid
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022
Animal models in endometriosis research
643
hormones as well as to drugs interfering with steroid action. Cor-
responding to the situation in humans, the growth of the ectopic
endometrial tissue in both rodent species was shown to be estro-
gen dependent (Vernon and Wilson, 1985; Rajkumar et al., 1990;
Rossi et al., 2000). In rats which had been ovariectomized 3 weeks
after engrafts of uterine tissue, the ectopic fragments recovered
much better in animals treated with estrogen alone after ovariec-
tomy than in those with combined estrogen and progesterone treat-
ment (Schor et al., 1999). Fang et al. (2004) confirmed the
important role for estrogen as a factor determining the size of the
implants in mice and demonstrated that the antiproliferative
effect of progesterone on the estrogen-dependent growth of
endometriotic tissue is mediated by intact progesterone receptor,
because this estrogen-dependent growth could be suppressed by
progesterone in uterine tissues of wild-type but not of progester-
one receptor-deficient mice.
According to the situation in women, the regression of uterine
ectopic implants could be induced in rats by generating a hypoes-
trogenic state, for example, by ovariectomy (Kudoh et al., 1997),
by the application of GnRH agonists (Sakata et al., 1990; Wright
and Sharpe-Timms, 1995; Kudoh et al., 1997), by synthetic pro-
gestational compounds such as levonorgestrel or dienogest (Jones,
1984; Katsuki et al., 1998) or by danazol treatment (Sakata et al.,
1990). Corresponding effects could be achieved in this model by
reducing the estrogen concentration by the application of anti-
estrogens (Saito et al., 2003), by using the selective estrogen
receptor modulator raloxifen (Yao et al., 2005) or by treatment
with aromatase inhibitors which interfere with estrogen synthesis
(Yano et al., 1996; Kudoh et al., 1997).
Moreover, the autologous rat model in particular has been
extensively used for studies of immune-modulating drugs and
anti-inflammatory agents in endometriosis. Uchiide et al. (2002)
demonstrated that uterine autotransplantation in rats induces the
infiltration of allergic inflammatory-related cells in peritoneal
stroma attached to the ectopic uterine tissue. Immunomodulation
by intraperitoneal or subcutaneous treatment with interferon-α-2b
in the rat reduced the size of induced lesions as monitored by con-
secutive laparotomies over a period of up to 4 months (Ingelmo
et al., 1999) as did the application of the immunomodulator
loxoribine in rats (Keenan et al., 1999) and the intraperitoneal
injection of interleukin-12 in a syngeneic mouse model
(Somigliana et al., 1999).
In addition, the development of experimentally induced
endometriosis in rats was inhibited by recombinant human
tumour necrosis factor (TNF)-binding protein-1 (r-hTBP-1),
which is the soluble form of TNF receptor type I (D’Antonio
et al., 2000). This effect was also seen after treatments of rats
with pentoxifylline, an anti-inflammatory substance reported to
reduce the production of inflammatory cytokines without induc-
ing a hypoestrogenic state (Nothnick et al., 1994), as well as
with ciglitazone, a ligand for proliferator-activated receptor-γ
(PPAR-γ) (Lebovic et al., 2004). Moreover, the initial develop-
ment of ectopic implants in rats has been reduced by cyclooxy-
genase-2 (COX-2) inhibitors (Matsuzaki et al., 2004) and also by
the application of various non-steroidal anti-inflammatory drugs
such as celecobix, indomethacin, sulinac and ibuprofen (but not
aspirin) in a mouse model for endometriosis (Efstathiou et al.,
2005).
Furthermore, this rodent model provides the opportunity to
investigate the effect of environmental contaminants on the estab-
lishment of ectopic uterine implants. It has been demonstrated that
exposure to dioxin preceding the surgical induction of endometri-
osis produces a dose-dependent increase in endometriotic site
diameter in rats and mice, which was much stronger in mice
(Cummings et al., 1996).
Recently, in an interesting approach, Dinulescu et al. (2005)
described the first mouse model of de-novo endometriosis. Activa-
tion of the oncogene K-ras in ovarian surface epithelial cells, but
not in cells of the peritoneal lining, resulted in the development of
benign epithelial lesions with histomorphological features of
human endometriosis. In addition to developing ovarian lesions,
nearly half the mice also developed peritoneal endometriosis 8
months after the K-ras activation of ovarian surface epithelial
cells. In combination with a conditional deletion of Pten, which
has been implicated in the progression of ovarian endometriosis to
endometrioid ovarian carcinoma in humans (Sato et al., 2000), K-
ras expression even induced metastatic endometrioid ovarian ade-
nocarcinomas. However, to date, no mutations of K-ras have been
found in human endometriosis (Vercellini et al., 1994; Otsuka
et al., 2004).
The hallmark symptoms of endometriosis in women include
severe pelvic pain and significantly reduced fertility (Elsheikh
et al., 2003). The influence of ectopic lesions on fertility has been
investigated in laboratory rodents. Although Cummings and
Metcalf (1996) did not observe a decrease in fertility in mice, a
reduced fecundity could be observed in rats with artificially
induced endometriosis (Vernon and Wilson, 1985), which could
be due in part to an increased number of luteinized unruptured
ovarian follicles (Moon et al., 1993). In addition, a role of pelvic
adhesions has not be excluded. Moreover, peritoneal inflamma-
tory cell hyperactivation could have an effect on fertility in
endometriosis. This was supported by the work of Steinleitner
et al. (1991a), demonstrating that macrophage-mediated subfertil-
ity in the mouse model could be compensated by pentoxifylline, a
drug that reverses the effect of macrophage hyperactivation, and
that the drug-mediated inhibition of macrophage activation
enhanced fertility in a hamster endometriosis model (Steinleitner
et al., 1991b).
Recently, studies have been published investigating the effect of
ectopic endometrial lesions on pain behaviour in the rat model of
endometriosis. Here, the autotransplanted ectopic endometrial
cystic fragments develop their own innervation including sympa-
thetic efferent as well as sensory fibres (Berkley et al., 2004),
which may have a general influence on the nervous system. This is
supported by the finding that vaginal nociception was increased in
rats with endometrial cysts similar to the situation in women with
endometriosis. In addition, those rats also displayed vaginal
hyperalgesia (Berkley et al., 2001), one of the symptoms of
increased pelvic pain in women. It is speculated that neuroactive
agents contained in the endometrial cysts may activate nociceptive
afferents influencing central neural mechanisms associated with
vaginal nociception as well as with reproduction via viscero–
visceral interactions (Cason et al., 2003; Berkley et al., 2004). It
remains to be elucidated whether the rats exhibit chronic pelvic
pain symptoms other than vaginal hyperalgesia in this model of
endometriosis.
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022
R.Grümmer
644
Evaluation of the homologous model
There are relevant differences between rodent and human repro-
ductive physiology; consequently, these models possess limita-
tions. Because rodents do not menstruate, they do not develop
spontaneous endometriosis, and the disease has to be induced
artificially by the autotransplantation of uterine tissue. In addi-
tion, in most of these rodent studies, the uterine fragments which
were transplanted included the myometrial layer which could
affect the development of the lesions. When evaluating the
weight and size of the ectopic lesions, the myometrium could
make up a considerable proportion, not representing the situation
of human endometriotic lesions.
Despite these limitations, the rodent model offers significant
advantages. Apart from the limited costs and the opportunity to
perform studies in large groups of genetically similar animals,
long-term studies can be performed because there is no rejection
of the autotransplanted ectopic tissue. It is well suited for the
investigation of mechanisms involved in the peritoneal attachment
of endometrial cells as well as of effects of therapeutical drugs and
chemicals, and it allows the evaluation of ectopic lesions at differ-
ent time intervals. Moreover, the homologous rodent model of sur-
gically induced endometriosis provides a well-characterized
immune system and thus is appropriate and extensively used to
study the effect of immune-modulating drugs and anti-inflamma-
tory agents on endometriosis. Furthermore, in regard to new
insights in the aetiology of endometriosis obtained by genomic
and proteomic approaches, transgenic mice models may gain
importance as models of endometriosis to examine functions
driven by single genes.
Xenotransplantation of human endometrium to immunodeficient
mice
Characterization of the heterologous model
Because causal factors for the development and maintenance of
endometriosis could originate in the endometrium itself and may
not be present in the rodent endometrium, models have been
developed by implanting human endometrial tissue instead of
autologous endometrium into immunodeficient mouse strains.
Most widely used for this purpose is the athymic nude mouse,
which lacks mature T lymphocytes. Human endometrial fragments
from the proliferative or secretory phase of the menstrual cycle
(Zamah et al., 1984; Bergqvist et al., 1985; Zaino et al., 1985;
Bruner et al., 1997; Nisolle et al., 2000; Grummer et al., 2001;
Beliard et al., 2002) as well as the menstrual endometrium
(Nisolle et al., 2000; Van Langendonckt et al., 2004; Eggermont
et al., 2005) have been successfully implanted either subcutane-
ously or intraperitoneally into nude mice. These fragments implant
and form endometriotic-like lesions which resemble those found
in patients in terms of macroscopic and histological appearance,
whereas the stage of the menstrual cycle at the time of collecting
the human tissue seems to have no impact on the development of
those ectopic lesions (Nisolle et al., 2000). Preservation of estro-
gen and progesterone receptors and steroid responsiveness have
been demonstrated in the ectopic human tissue (Zamah et al.,
1984; Bruner et al., 1997, 1999; Grummer et al., 2001). Angio-
genesis guarantees the maintenance of the transplants and the
transport of systemically applied drugs to the human endometrial
tissue. The establishment of blood vessels of mouse origin occurred
from just 4 days after transplantation onwards, independent of the
localization of the ectopic lesions (Grummer et al., 2001; Hull
et al., 2003).
When endometrial tissue was randomly inoculated into the peri-
toneal cavity, peritoneal adhesion occurred from 2 days after
implantation onwards. The sites of implantation of the endometrial
fragments were preferentially the gut, the muscles of the abdominal
wall, the liver and the adipose fat surrounding abdominal organs
(Grummer et al., 2001; Fortin et al., 2003). However, studies from
our group as well as others revealed that the recovery rate is highly
variable from one animal to another and in average does not exceed
30% (Bruner et al., 1997; Grummer et al., 2001). This ‘take-rate’ is
further reduced after in vitro culturing of endometrial tissue for 24
h before transplantation but could be enhanced to 100% by suturing
the human endometrial fragments to different intraperitoneal sites
(Grummer et al., 2001). This is favourable, in regard to drug test-
ing, because a difference in the number of lesions in the two groups
of animals could be due to different ‘take-rates’ at the beginning of
the experiment rather than to the effect of a drug.
For non-invasive detection and quantification of implanted
endometrial tissue, Tabibzadeh et al. (1999) loaded dissociated
glands and stromal cells with a lipophylic fluorescent dye (DiO)
before in vivo transplantation. However, an effect of the solvent
[dimethylsulphoxide (DMSO) and absolute ethanol] on the
endometrial tissue cannot be excluded. An interesting approach
that is better for monitoring of subcutaneously or intraperitoneally
transplanted endometrial fragments was recently described by
Fortin et al. (2003). They transduced endometrial fragments of the
proliferative phase with GFP cDNA before implantation, resulting
in a transient fluorescence of the endometrial tissue allowing opti-
cal in vivo imaging of the implantation and development of
ectopic lesions. Because adenoviruses are transient expression
vectors, they observed that fluorescence of endometrial tissue
fades out around the third or fourth week, limiting the non-inva-
sive follow-up to this time frame. In addition, when using GFP as
a reporter gene, in vivo quantification would probably be limited
to subcutaneous lesions, which can be directly visualized through
the skin of living mice, because the wavelength of GFP-emitted
light from intraperitoneal fragments is absorbed by surrounding
tissues. Thus, this model allows dynamic studies of lesion by mon-
itoring implantation and development. However, a yet unknown
interaction between GFP and lesions or applicated drugs might
interfere with the studies.
Because, in the heterologous mouse model, human tissue is
transplanted, the time frame for maintenance of intact and well-
preserved tissue is limited. In most of the studies performed, cul-
ture of human endometrium in nude mice does not exceed a period
of 4 weeks. From 3 weeks after inoculation onwards, dedifferenti-
ation parameters and lymphocyte infiltration can be observed
(Grummer et al., 2001).
Because longer maintenance of human tissue in this animal
model would be advantageous for special experimental
approaches, mice with an even more defective immune system
have been established as heterologous models for endometriosis.
Human endometrial tissue has successfully been transplanted to
severe combined immunodeficient (SCID) mice (Aoki et al.,
1994; Kaufmann et al., 1995; Awwad et al., 1999) as well as to
non-obese diabetic (NOD)-SCID mice (Grummer et al., 2001),
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022
Animal models in endometriosis research
645
both of which show a combined congenital deficiency in T- and B-
lymphocyte function. Compared with the nude mouse model,
human endometrium engrafted in SCID/NOD-SCID mice
revealed a higher ‘take-rate’ of fragments (Aoki et al., 1994) and a
better preserved morphology and expression of steroid hormone
receptors over a period of 4 weeks (Grummer et al., 2001).
Although these mice lack B and T lymphocytes, they do have
some natural killer (NK) cell activity. Recently, Matsuura-Sawada
et al. (2005) cultured human endometrial tissue subcutaneously in
NOD/SCID/γ Cnull
(NOG) immunodeficient mice which are addi-
tionally defective in NK-cell activity. In this model, they repro-
duced one human menstrual cycle of 28 days by hormone
application in ovariectomized animals. At the level of histology,
they showed cyclic changes also observed in eutopic human
endometrium but without signs of decidualization.
Greenberg and Slayden (2004) subcutaneously engrafted
human endometriotic tissue in transgenic recombinase-activating
gene-2 knockout [RAG-2/γ(c)KO] mice, which are not only
devoid of B and T lymphocytes but also lack NK cells. Only
endometriotic biopsy specimens that contained endometrium-like
glands developed visible lesions. Red peritoneal lesions, but not
biopsy specimens of ovarian endometrioma, established grafts at a
high rate. In this animal model for endometriosis, they mimicked
four consecutive human menstrual cycles by applying estrogen
and progesterone via hormone-releasing capsules. The established
grafts were well-vascularized and maintained steroid hormone
receptors.
Application of the heterologous model
Animal models of endometriosis based on the xenotransplantation
of human endometrium permit the elucidation of mechanisms
involved in the establishment of endometriosis as well as testing
the effects of potential therapeutical compounds on human tissue.
One approach to prevent the development of peritoneal endometri-
osis could target the adherence of endometrial tissue to and the
invasion of the peritoneal lining. MMPs are involved in such inva-
sion processes (Hudelist et al., 2005; Zhou and Nothnick, 2005). It
could be shown that MMPs are regulated by steroid hormones and
cytokines in endometrial tissue and that the suppression of MMPs
inhibits the establishment of ectopic lesions by human
endometrium in nude mice (Bruner et al., 1997, 1999; Bruner-
Tran et al., 2002).
Another therapeutical target discussed is the development of
new blood vessels, which is essential for the persistence of ectopic
endometriotic lesions. Endometrial xenografts in nude mice have
been shown to produce high levels of VEGF (Nisolle et al., 2000),
and anti-angiogenic agents such as soluble VEGF receptor (sflt-1)
and VEGF-A-antibodies significantly inhibited the growth of
endometriotic lesions in nude mice (Hull et al., 2003).
Because endometriosis is an estrogen-dependent disease, the
responsiveness of human ectopic lesions to steroid hormones has
been investigated in this mouse model. Pretreatment of human
endometrial cells with estrogen alone or in combination with a
progestin, but not with progestin alone, before injection into the
peritoneal cavity of nude mice resulted in a higher percentage of
animals developing endometriotic-like lesions (Beliard et al.,
2002). Using optical in vivo imaging, Fortin et al. (2004) demon-
strated a significantly reduced lesion size in estrogen-untreated
versus estrogen-treated ovariectomized nude mice.
Recent studies have identified aberrations in the expression of
aromatase and 17β-hydroxysteroid reductase-2 in human endome-
triotic lesions (Zeitoun et al., 1998; Bulun et al., 1999; Zeitoun
and Bulun, 1999), suggesting a role for steroid-metabolizing
enzymes in the pathogenesis of endometriosis. We could demon-
strate that the transcription of estrogen-metabolizing enzymes is
maintained in human endometrial tissue cultured in nude mice and
is regulated by the exogenously applied drugs danazol, dydroges-
terone and medroxyprogesterone acetate, which are used in the
treatment of endometriosis and are known to interact with the
estrogen metabolism, as well as by an aromatase inhibitor
(Gruemmer et al., 2005).
In contrast to the homologous rodent model, mice used in the
heterologous models have a deficient immune system and thus are
hardly used in studies evaluating the effect of immune-modulating
drugs. Application of an inhibitor of COX-2, which is known to be
up-regulated at sites of inflammation, had no influence on the
number or size of ectopic endometrial lesions in nude mice (Hull
et al., 2005). However, anti-inflammatory components, contained
for example in peritoneal fluid of women, could be investigated in
this model. Tabibzadeh et al. (1999) demonstrated that endome-
trial cells suspended in peritoneal fluid of women without
endometriosis established significantly less ectopic lesions than
those suspended in peritoneal fluid of women with endometriosis
after intraperitoneal injection into nude mice.
Evaluation of the heterologous model
This model shares the advantages described for the homologous
model, such as easy availability and low costs, as well as some of
the limitations such as differences between rodent and human
physiology. However, it does not provide an intact immune sys-
tem. Yet, the heterologous mouse model for endometriosis repre-
sents a promising tool for experimental approaches evaluating the
aetiology of this disease and for therapeutic testing of pharmacolog-
ical and hormonal modulations because it is based on human
endometrium. For these investigations, human endometrial tissue
can be taken from women with and without endometriosis as well as
from endometriotic lesions. Regulatory mechanisms, for example,
after drug administration, can be evaluated in the human ectopic tis-
sue in an in vivo situation. Long-term studies that are not feasible in
women can be performed, and the hormonal status of the human
menstrual cycle can be mimicked in ovariectomized animals.
Recently established methods using fluorescence allow in vivo
imaging and may improve the quantification of both size and
number of the ectopic lesions. In addition, techniques using mag-
netic resonance imaging (MRI) for in vivo monitoring of the
lesions are in progress.
Primate models
Characterization of the primate model
Menstruating primates develop spontaneous endometriosis, estab-
lishing ectopic lesions histologically identical and at similar sites
compared with the human disease (MacKenzie and Casey, 1975;
D’Hooghe et al., 1991; Dick et al., 2003). Spontaneous endometri-
osis does not develop with a high frequency in all monkeys but
could be shown to increase with time in capture possibly because
of less pregnancies leading to a higher number of consecutive
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022
R.Grümmer
646
menstrual cycles with increased exposure to retrograde menstrua-
tion (D’Hooghe et al., 1996a). Although spontaneous endometrio-
sis has been reported for 11 species of non-human primates (for
review, see Story and Kennedy, 2004), most studies have been
performed in rhesus macaques and baboons.
Because spontaneous endometriosis in monkeys develops
slowly over a period of several years, methods have been
employed for the artificial induction of this disease. In the first
studies, menstrual flow was diverted into the abdomen by reposi-
tioning the cervix of rhesus monkeys (Te Linde and Scott, 1950)
and later by occluding the cervix to increase the amount of retro-
grade menstruation in baboons (D’Hooghe et al., 1994). In addi-
tion, surgical induction of the disease can be performed by
suturing fragments of endometrial tissue at ectopic sites or by
seeding the peritoneal cavity with fragments of endometrial tissue
(D’Hooghe et al., 1995b; Yang et al., 2000; Fazleabas et al.,
2002). D’Hooghe et al. (1995b) induced endometriosis in a
baboon model by intraperitoneal injection of endometrial tissue,
demonstrating that endometriosis developed similarly to that
observed in the spontaneous disease and that ectopic lesions were
induced more efficiently by inoculating menstrual endometrium
compared with luteal phase endometrium. Although, in this study,
menstrual endometrium was extracted from each baboon by uter-
ine curettage, thus also containing endometrial cell layers not
physiologically shed during menstruation, other studies used shed
menstrual endometrium obtained using a pipelle (Fazleabas et al.,
2002, 2003). By this means, induced ectopic endometrial lesions
in the baboon could be shown to express estrogen receptor-β,
MMP-7, VEGF and aromatase (Fazleabas et al., 2002, 2003).
Ongoing efforts focus on the non-invasive diagnosis for the identifi-
cation of intraperitoneal lesions using MRI (Zondervan et al., 2004).
These methods of induced endometriosis in non-human pri-
mates have been extensively reviewed previously (D’Hooghe,
1997; D’Hooghe and Debrock, 2002; Story and Kennedy, 2004;
Fazleabas, 2005).
Application of the primate model
Many studies have been performed evaluating spontaneous
endometriosis on the one hand and using artificially induced
endometriosis on the other hand.
It could be shown that elevated estrogen levels as well as fre-
quent hysterotomies, but not laparoscopies, lead to an increased
risk for the development of spontaneous endometriosis (Hadfield
et al., 1997) and that genetic predisposition also seems to contrib-
ute to a higher risk for this disease (Zondervan et al., 2004).
Regression of peritoneal endometriotic lesions could be demon-
strated after the application of progesterone receptor modulators,
either alone or in combination with a GnRH analogue, in induced
endometriosis in cynomologous monkeys (Grow et al., 1996). The
influence of environmental compounds on the development of
spontaneous endometriosis was proven by exposure of rhesus
monkeys to dioxin or polychlorinated biphenyls (PCBs), which
resulted in a dose-dependent increase in incidence and severity of
endometriosis (Rier et al., 1993; Vanden Heuvel et al., 1994;
DeVito et al., 1995).
The primate model is well suited to investigate the role of the
immune system on the development and progression of endome-
triosis. Subclinical peritoneal inflammation occurs in baboons
during menstruation as well as after the intrapelvic injection of
endometrium, as demonstrated by an increase in peritoneal fluid
leukocyte concentration and the amount of peritoneal fluid cells
positive for TNF-α, transforming growth factor (TGF-β1) and cer-
tain leukocyte markers (D’Hooghe et al., 2001). It was shown that
immunosuppression led to an increase in the number and size of
spontaneously developed endometriotic lesions (D’Hooghe et al.,
1995a). Neutralization of TNF-α with r-hTBP-1 inhibits the
development of endometriotic lesions and adhesions after the arti-
ficial induction of the disease (D’Hooghe et al., 2006), and a
decrease in active red lesion number and surface area was
observed in female baboons with spontaneous peritoneal endome-
triosis after neutralizing TNF activity by treatment with etanercept
(Barrier et al., 2004).
With regard to the impact of endometriosis on fertility, it was
proven that mild-to-severe endometriosis leads to lower preg-
nancy rates in baboons (D’Hooghe et al., 1996b) as well as to a
reduction in chemical and term pregnancy rates in another monkey
species, Macaca fascicularis (Schenken et al., 1984). Whether
these effects on fertility are due to peritoneal adhesions and/or to
immunological responses is still to be investigated. In addition, a
feedback effect of the endometriotic lesions on the eutopic
endometrium is discussed, and it has to be evaluated whether the
differences in fecundity result in, or are the result of, endometrio-
sis. Whether the peritoneal presence of ectopic implantation sites
influences the physiology of the eutopic endometrium can effec-
tively be investigated in the primate model. Recently, an increase
in the angiogenic factor Cyr61, which is known to be deregulated
in the endometrium of women with endometriosis (Absenger
et al., 2004), could be demonstrated in the eutopic endometrium of
baboons following peritoneal inoculation with menstrual
endometrium (Gashaw et al., 2006). This study provides evidence
for a feedback mechanism from established lesions to induce a
shift in gene expression patterns in the eutopic endometrium in the
baboon.
Evaluation of the primate model
There is no doubt that primate models for endometriosis are most
closely related to the situation in humans and thus are most appro-
priate to investigate the numerous aspects of this disease, includ-
ing the initiation and progression of endometriosis, immunological
parameters, inheritance patterns as well as feedback mechanisms
on the eutopic endometrium. They reveal many similarities to
women with respect to pelvic anatomy, reproductive physiological
characteristics and immunological features. Primates develop
endometriosis spontaneously, and the disease can also be induced
for research purposes. They represent the only experimental
model, allowing the comparison of parameters in spontaneous
endometriosis with parameters before and after disease induction,
thus gaining information about cause and effect of the disease.
However, ethical considerations and particularly the high costs of
performing experiments using primates limit the use of this model
in endometriosis research.
Conclusions
Animal models of endometriosis are of extreme value and indis-
pensable for the evaluation of pathophysiological mechanisms
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022
Animal models in endometriosis research
647
underlying the development of this prevalent gynaecological dis-
ease. This review summarizes different commonly used animal
models, their potentials limitations. These have to be weighted up
when designing experimental approaches with regard to their
applicability, for example, considering species-specific character-
istics of metabolism or reproductive physiology, as well as to their
cost–effect ratio. New insights into the aetiology of endometriosis
will be obtained using genomic and proteomic approaches. In this
regard, transgenic mice models might gain importance as models of
endometriosis to examine functions driven by single genes. In gen-
eral, animal models will help to develop novel non-invasive diag-
nostic tools and improved therapeutical approaches for a better
treatment of endometriosis in women.
References
Absenger Y, Hess-Stumpp H, Kreft B, Kratzschmar J, Haendler B, Schutze N,
Regidor PA and Winterhager E (2004) Cyr61, a deregulated gene in
endometriosis. Mol Hum Reprod 10,399–407.
Aoki D, Katsuki Y, Shimizu A, Kakinuma C and Nozawa S (1994) Successful
heterotransplantation of human endometrium in SCID mice. Obstet Gyne-
col 83,220–228.
Armstrong PB, Quigley JP and Sidebottom E (1982) Transepithelial invasion
and intramesenchymal infiltration of the chick embryo chorioallantois by
tumor cell lines. Cancer Res 42,1826–1837.
Awwad JT, Sayegh RA, Tao XJ, Hassan T, Awwad ST and Isaacson K (1999)
The SCID mouse: an experimental model for endometriosis. Hum Reprod
14,3107–3111.
Barrier BF, Bates GW, Leland MM, Leach DA, Robinson RD and Propst AM
(2004) Efficacy of anti-tumor necrosis factor therapy in the treatment of
spontaneous endometriosis in baboons. Fertil Steril 81,775–779.
Beliard A, Noel A, Goffin F, Frankenne F and Foidart JM (2002) Role of endo-
crine status and cell type in adhesion of human endometrial cells to the
peritoneum in nude mice. Fertil Steril 78,973–978.
Bergqvist A, Jeppsson S, Kullander S and Ljungberg O (1985) Human uterine
endometrium and endometriotic tissue transplanted into nude mice. Mor-
phologic effects of various steroid hormones. Am J Pathol 121,337–341.
Berkley KJ, Cason A, Jacobs H, Bradshaw H and Wood E (2001) Vaginal
hyperalgesia in a rat model of endometriosis. Neurosci Lett 306,185–188.
Berkley KJ, Dmitrieva N, Curtis KS and Papka RE (2004) Innervation of
ectopic endometrium in a rat model of endometriosis. Proc Natl Acad Sci
USA 101,11094–11098.
Bruner KL, Matrisian LM, Rodgers WH, Gorstein F and Osteen KG (1997)
Suppression of matrix metalloproteinases inhibits establishment of ectopic
lesions by human endometrium in nude mice. J Clin Invest 99,2851–2857.
Bruner KL, Eisenberg E, Gorstein F and Osteen KG (1999) Progesterone and
transforming growth factor-beta coordinately regulate suppression of
endometrial matrix metalloproteinases in a model of experimental
endometriosis. Steroids 64,648–653.
Bruner-Tran KL, Eisenberg E, Yeaman GR, Anderson TA, McBean J and
Osteen KG (2002) Steroid and cytokine regulation of matrix metallopro-
teinase expression in endometriosis and the establishment of experimental
endometriosis in nude mice. J Clin Endocrinol Metab 87,4782–4791.
Bulun SE, Zeitoun K, Takayama K, Noble L, Michael D, Simpson E, Johns A,
Putman M and Sasano H (1999) Estrogen production in endometriosis and
use of aromatase inhibitors to treat endometriosis. Endocr Relat Cancer
6,293–301.
Cason AM, Samuelsen CL and Berkley KJ (2003) Estrous changes in vaginal
nociception in a rat model of endometriosis. Horm Behav 44,123–131.
Cummings AM and Metcalf JL (1995) Induction of endometriosis in mice: a
new model sensitive to estrogen. Reprod Toxicol 9,233–238.
Cummings AM and Metcalf JL (1996) Effect of surgically induced endometri-
osis on pregnancy and effect of pregnancy and lactation on endometriosis
in mice. Proc Soc Exp Biol Med 212,332–337.
Cummings AM, Metcalf JL and Birnbaum L (1996) Promotion of endome-
triosis by 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats and mice: time-
dose dependence and species comparison. Toxicol Appl Pharmacol
138,131–139.
D’Antonio M, Martelli F, Peano S, Papoian R and Borrelli F (2000) Ability of
recombinant human TNF binding protein-1 (r-hTBP-1) to inhibit the
development of experimentally-induced endometriosis in rats. J Reprod
Immunol 48,81–98.
DeVito MJ, Birnbaum LS, Farland WH and Gasiewicz TA (1995) Compari-
sons of estimated human body burdens of dioxinlike chemicals and TCDD
body burdens in experimentally exposed animals. Environ Health Per-
spect 103,820–831.
D’Hooghe TM (1997) Clinical relevance of the baboon as a model for the
study of endometriosis. Fertil Steril 68,613–625.
D’Hooghe TM and Debrock S (2002) Endometriosis, retrograde menstruation
and peritoneal inflammation in women and in baboons. Hum Reprod
Update 8,84–88.
D’Hooghe TM, Bambra CS, Cornillie FJ, Isahakia M and Koninckx PR (1991)
Prevalence and laparoscopic appearance of spontaneous endometriosis in
the baboon (Papio anubis, Papio cynocephalus). Biol Reprod 45,411–416.
D’Hooghe TM, Bambra CS, Suleman MA, Dunselman GA, Evers HL and
Koninckx PR (1994) Development of a model of retrograde menstruation
in baboons (Papio anubis). Fertil Steril 62,635–638.
D’Hooghe TM, Bambra CS, Raeymaekers BM, De Jonge I, Hill JA and
Koninckx PR (1995a) The effects of immunosuppression on development
and progression of endometriosis in baboons (Papio anubis). Fertil Steril
64,172–178.
D’Hooghe TM, Bambra CS, Raeymaekers BM, De Jonge I, Lauweryns JM
and Koninckx PR (1995b) Intrapelvic injection of menstrual endometrium
causes endometriosis in baboons (Papio cynocephalus and Papio anubis).
Am J Obstet Gynecol 173,125–134.
D’Hooghe TM, Bambra CS, De Jonge I, Lauweryns JM and Koninckx PR
(1996a) The prevalence of spontaneous endometriosis in the baboon
(Papio anubis, Papio cynocephalus) increases with the duration of captiv-
ity. Acta Obstet Gynecol Scand 75,98–101.
D’Hooghe TM, Bambra CS, Raeymaekers BM, Riday AM, Suleman MA and
Koninckx PR (1996b) The cycle pregnancy rate is normal in baboons with
stage I endometriosis but decreased in primates with stage II and stage III–
IV disease. Fertil Steril 66,809–813.
D’Hooghe TM, Bambra CS, Xiao L, Peixe K and Hill JA (2001) Effect of
menstruation and intrapelvic injection of endometrium on inflammatory
parameters of peritoneal fluid in the baboon (Papio anubis and Papio
cynocephalus). Am J Obstet Gynecol 184,917–925.
D’Hooghe TM, Nugent NP, Cuneo S, Chai DC, Deer F, Debrock S, Kyama
CM, Mihalyi A and Mwenda JM (2006) Recombinant human TNFRSF1A
(r-hTBP1) inhibits the development of endometriosis in baboons: a pro-
spective, randomized, placebo- and drug-controlled study. Biol Reprod
74,131–136.
Dick EJ Jr, Hubbard GB, Martin LJ and Leland MM (2003) Record review of
baboons with histologically confirmed endometriosis in a large estab-
lished colony. J Med Primatol 32,39–47.
Dinulescu DM, Ince TA, Quade BJ, Shafer SA, Crowley D and Jacks T (2005)
Role of K-ras and Pten in the development of mouse models of endometri-
osis and endometrioid ovarian cancer. Nat Med 11,63–70.
Dunselman GA, Willebrand D, Land JA, Bouckaert PX and Evers JL (1989) A
rabbit model of endometriosis. Gynecol Obstet Invest 27,29–33.
Efstathiou JA, Sampson DA, Levine Z, Rohan RM, Zurakowski D, Folkman J,
D’Amato RJ and Rupnick MA (2005) Nonsteroidal antiinflammatory
drugs differentially suppress endometriosis in a murine model. Fertil
Steril 83,171–181.
Eggermont J, Donnez J, Casanas-Roux F, Scholtes H and Van Langendonckt A
(2005) Time course of pelvic endometriotic lesion revascularization in a
nude mouse model. Fertil Steril 84,492–499.
Elsheikh A, Milingos S, Loutradis D, Kallipolitis G and Michalas S (2003)
Endometriosis and reproductive disorders. Ann N Y Acad Sci 997,247–254.
Fang Z, Yang S, Lydon JP, DeMayo F, Tamura M, Gurates B and Bulun SE
(2004) Intact progesterone receptors are essential to counteract the prolif-
erative effect of estradiol in a genetically engineered mouse model of
endometriosis. Fertil Steril 82,673–678.
Fazleabas AT (2005) A baboon model for inducing endometriosis. Methods
Mol Med 121,93–98.
Fazleabas AT, Brudney A, Gurates B, Chai D and Bulun S (2002) A modified
baboon model for endometriosis. Ann N Y Acad Sci 955,308–317; discus-
sion 340–342, 396–406.
Fazleabas AT, Brudney A, Chai D, Langoi D and Bulun SE (2003) Steroid
receptor and aromatase expression in baboon endometriotic lesions. Fertil
Steril 80(Suppl 2),820–827.
Fortin M, Lepine M, Page M, Osteen K, Massie B, Hugo P and Steff AM
(2003) An improved mouse model for endometriosis allows noninvasive
assessment of lesion implantation and development. Fertil Steril 80(Suppl
2),832–838.
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022
R.Grümmer
648
Fortin M, Lepine M, Merlen Y, Thibeault I, Rancourt C, Gosselin D, Hugo P
and Steff AM (2004) Quantitative assessment of human endometriotic tis-
sue maintenance and regression in a noninvasive mouse model of
endometriosis. Mol Ther 9,540–547.
Gashaw I, Hastings JM, Jackson KS, Winterhager E and Fazleabas AT (2006)
Induced endometriosis in the baboon (Papio anubis) increases the expres-
sion of the proangiogenic factor Cyr61 (CCN1) in eutopic and ectopic
endometria. Biol Reprod 74,1060–1066.
Gescher DM, Siggelkow W, Meyhoefer-Malik A and Malik E (2005) A priori
implantation potential does not differ in eutopic endometrium of patients
with and without endometriosis. Arch Gynecol Obstet 272,117–123.
Giannopoulou E, Katsoris P, Hatziapostolou M, Kardamakis D, Kotsaki E,
Polytarchou C, Parthymou A, Papaioannou S and Papadimitriou E (2001)
X-rays modulate extracellular matrix in vivo. Int J Cancer 94,690–698.
Giudice LC and Kao LC (2004) Endometriosis. Lancet 364,1789–1799.
Golan A, Winston RM and Dargenio R (1984) Experimental endometriosis: a
microsurgical animal model in rats. Isr J Med Sci 20,1094–1096.
Greenberg LH and Slayden OD (2004) Human endometriotic xenografts in
immunodeficient RAG-2/gamma(c)KO mice. Am J Obstet Gynecol
190,1788–1795; discussion 1795–1796.
Grow DR, Williams RF, Hsiu JG and Hodgen GD (1996) Antiprogestin and/or
gonadotropin-releasing hormone agonist for endometriosis treatment and
bone maintenance: a 1-year primate study. J Clin Endocrinol Metab
81,1933–1939.
Grummer R, Schwarzer F, Bainczyk K, Hess-Stumpp H, Regidor PA, Schindler
AE and Winterhager E (2001) Peritoneal endometriosis: validation of an
in-vivo model. Hum Reprod 16,1736–1743.
Gruemmer R, Fechner S, Schwarzer F, Schmidt M and Winterhager E (2005)
Expression of angiogenic factors and regulation of estrogen metabolising
enzymes in human endometrial lesions grown in nude mice. Eur J Obstet
Gynecol Reprod Med 123,S56 (abstract).
Hadfield RM, Yudkin PL, Coe CL, Scheffler J, Uno H, Barlow DH, Kemnitz
JW and Kennedy SH (1997) Risk factors for endometriosis in the rhesus
monkey (Macaca mulatta): a case–control study. Hum Reprod Update
3,109–115.
Halme J, Hammond MG, Hulka JF, Raj SG and Talbert LM (1984) Retrograde
menstruation in healthy women and in patients with endometriosis. Obstet
Gynecol 64,151–154.
Hirata T, Osuga Y, Yoshino O, Hirota Y, Harada M, Takemura Y, Morimoto C,
Koga K, Yano T, Tsutsumi O et al. (2005) Development of an experimen-
tal model of endometriosis using mice that ubiquitously express green flu-
orescent protein. Hum Reprod 20,2092–2096.
Homm RJ, Garza DE, Mathur S, Austin M, Baggett B and Williamson HO
(1989) Immunological aspects of surgically induced experimental
endometriosis: variation in response to therapy. Fertil Steril 52,132–139.
Hudelist G, Lass H, Keckstein J, Walter I, Wieser F, Wenzl R, Mueller R,
Czerwenka K, Kubista E and Singer CF (2005) Interleukin 1alpha and tissue-
lytic matrix metalloproteinase-1 are elevated in ectopic endometrium of
patients with endometriosis. Hum Reprod 20,1695–1701.
Hull ML, Charnock-Jones DS, Chan CL, Bruner-Tran KL, Osteen KG, Tom
BD, Fan TP and Smith SK (2003) Antiangiogenic agents are effective
inhibitors of endometriosis. J Clin Endocrinol Metab 88,2889–2899.
Hull ML, Prentice A, Wang DY, Butt RP, Phillips SC, Smith SK and Charnock-
Jones DS (2005) Nimesulide, a COX-2 inhibitor, does not reduce lesion
size or number in a nude mouse model of endometriosis. Hum Reprod
20,350–358.
Ingelmo JM, Quereda F and Acien P (1999) Intraperitoneal and subcutaneous
treatment of experimental endometriosis with recombinant human interferon-
alpha-2b in a murine model. Fertil Steril 71,907–911.
Jones RC (1984) The effect of a luteinizing hormone releasing hormone (LRH)
agonist (Wy-40,972), levonorgestrel, danazol and ovariectomy on experi-
mental endometriosis in the rat. Acta Endocrinol (Copenh) 106,282–288.
Katsuki Y, Takano Y, Futamura Y, Shibutani Y, Aoki D, Udagawa Y and
Nozawa S (1998) Effects of dienogest, a synthetic steroid, on experimen-
tal endometriosis in rats. Eur J Endocrinol 138,216–226.
Kaufmann R, Rudolphi A, Boxberger HJ, Hainzl A, Rosenthal H and Reimann J
(1995) Stable engraftment of human female genital mucous membrane
xenografts on SCID mice. Gynecol Obstet Invest 40,97–100.
Keenan JA, Williams-Boyce PK, Massey PJ, Chen TT, Caudle MR and
Bukovsky A (1999) Regression of endometrial explants in a rat model of
endometriosis treated with the immune modulators loxoribine and levami-
sole. Fertil Steril 72,135–141.
Kudoh M, Susaki Y, Ideyama Y, Nanya T, Mori M and Shikama H (1997)
Inhibitory effects of a novel aromatase inhibitor, YM511, on growth of
endometrial explants and insulin-like growth factor-I gene expression
in rats with experimental endometriosis. J Steroid Biochem Mol Biol
63,75–80.
Lapp T (2000) ACOG issues recommendations for the management of
endometriosis. American College of Obstetricians and Gynecologists. Am
Fam Physician 62,1431,1434.
Lebovic DI, Kir M and Casey CL (2004) Peroxisome proliferator-activated
receptor-gamma induces regression of endometrial explants in a rat model
of endometriosis. Fertil Steril 82(Suppl 3),1008–1013.
Maas JW, Groothuis PG, Dunselman GA, de Goeij AF, Struijker-Boudier HA
and Evers JL (2001) Development of endometriosis-like lesions after
transplantation of human endometrial fragments onto the chick embryo
chorioallantoic membrane. Hum Reprod 16,627–631.
MacKenzie WF and Casey HW (1975) Animal model of human disease.
Endometriosis. Animal model: endometriosis in rhesus monkeys. Am J
Pathol 80,341–344.
Malik E, Meyhofer-Malik A, Berg C, Bohm W, Kunzi-Rapp K, Diedrich K
and Ruck A (2000) Fluorescence diagnosis of endometriosis on the
chorioallantoic membrane using 5-aminolaevulinic acid. Hum Reprod
15,584–588.
Matsuura-Sawada R, Murakami T, Ozawa Y, Nabeshima H, Akahira J, Sato Y,
Koyanagi Y, Ito M, Terada Y and Okamura K (2005) Reproduction of
menstrual changes in transplanted human endometrial tissue in immuno-
deficient mice. Hum Reprod 20,1477–1484.
Matsuzaki S, Canis M, Darcha C, Dallel R, Okamura K and Mage G (2004)
Cyclooxygenase-2 selective inhibitor prevents implantation of eutopic
endometrium to ectopic sites in rats. Fertil Steril 82,1609–1615.
Moon CE, Bertero MC, Curry TE, London SN, Muse KN, Sharpe KL and
Vernon MW (1993) The presence of luteinized unruptured follicle syn-
drome and altered folliculogenesis in rats with surgically induced
endometriosis. Am J Obstet Gynecol 169,676–682.
Nap AW, Groothuis PG, Demir AY, Maas JW, Dunselman GA, de Goeij AF
and Evers JL (2003) Tissue integrity is essential for ectopic implantation
of human endometrium in the chicken chorioallantoic membrane. Hum
Reprod 18,30–34.
Nap AW, Dunselman GA, de Goeij AF, Evers JL and Groothuis PG
(2004) Inhibiting MMP activity prevents the development of endome-
triosis in the chicken chorioallantoic membrane model. Hum Reprod
19,2180–2187.
Nap AW, Dunselman GA, Griffioen AW, Mayo KH, Evers JL, Groothuis PG
(2005) Angiostatic agents prevent the development of endometriosis-like
lesions in the chicken chorioallantoic membrane. Fertil Steril 83,793–795.
Nisolle M, Casanas-Roux F and Donnez J (2000) Early-stage endometriosis:
adhesion and growth of human menstrual endometrium in nude mice. Fer-
til Steril 74,306–312.
Nothnick WB, Curry TE and Vernon MW (1994) Immunomodulation of rat
endometriotic implant growth and protein production. Am J Reprod
Immunol 31,151–162.
Oosterlynck DJ, Meuleman C, Waer M, Koninckx PR and Vandeputte M
(1993) Immunosuppressive activity of peritoneal fluid in women with
endometriosis. Obstet Gynecol 82,206–212.
Otsuka J, Okuda T, Sekizawa A, Amemiya S, Saito H, Okai T, Kushima M and
Tachikawa T (2004) K-ras mutation may promote carcinogenesis of
endometriosis leading to ovarian clear cell carcinoma. Med Electron
Microsc 37,188–192.
Pellicer A, Navarro J, Bosch E, Garrido N, Garcia-Velasco JA, Remohi J and
Simon C (2001) Endometrial quality in infertile women with endometrio-
sis. Ann N Y Acad Sci 943,122–130.
Rajkumar K, Schott PW and Simpson CW (1990) The rat as an animal model
for endometriosis to examine recurrence of ectopic endometrial tissue
after regression. Fertil Steril 53,921–925.
Rice VM (2002) Conventional medical therapies for endometriosis. Ann N Y
Acad Sci 955,343–352; discussion 389–393, 396–406.
Rier SE, Martin DC, Bowman RE, Dmowski WP and Becker JL (1993)
Endometriosis in rhesus monkeys (Macaca mulatta) following chronic
exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Fundam Appl Toxicol
21,433–441.
Rock JA, Prendergast RA, Bobbie D, Green WR, Parmley TH and Dubin NH
(1993) Intraocular endometrium in the rabbit as a model for endometrio-
sis. Fertil Steril 59,232–235.
Rossi G, Somigliana E, Moschetta M, Santorsola R, Cozzolino S, Filardo P,
Salmaso A and Zingrillo B (2000) Dynamic aspects of endometriosis in a
mouse model through analysis of implantation and progression. Arch
Gynecol Obstet 263,102–107.
Saito T, Yoshizawa M, Yamauchi Y, Kinoshita S, Fujii T, Mieda M, Sone H,
Yamamoto Y and Koizumi N (2003) Effects of the novel orally active
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022
Animal models in endometriosis research
649
antiestrogen TZE-5323 on experimental endometriosis. Arzneimittelfor-
schung 53,507–514.
Sakata M, Terakawa N, Mizutani T, Tanizawa O, Matsumoto K, Terada N and
Sudo K (1990) Effects of danazol, gonadotropin-releasing hormone agonist,
and a combination of danazol and gonadotropin-releasing hormone agonist
on experimental endometriosis. Am J Obstet Gynecol 163,1679–1684.
Sampson JA (1927) Perotoneal endometriosis due to the menstrual dissemina-
tion of endometrial tissue into the peritoneal cavity. Am J Obstet Gynecol
14,422–425.
Sato N, Tsunoda H, Nishida M, Morishita Y, Takimoto Y, Kubo T and
Noguchi M (2000) Loss of heterozygosity on 10q23.3 and mutation of
the tumor suppressor gene PTEN in benign endometrial cyst of the
ovary: possible sequence progression from benign endometrial cyst to
endometrioid carcinoma and clear cell carcinoma of the ovary. Cancer
Res 60,7052–7056.
Schenken RS and Asch RH (1980) Surgical induction of endometriosis in the
rabbit: effects on fertility and concentrations of peritoneal fluid prostag-
landins. Fertil Steril 34,581–587.
Schenken RS, Asch RH, Williams RF and Hodgen GD (1984) Etiology of
infertility in monkeys with endometriosis: luteinized unruptured follicles,
luteal phase defects, pelvic adhesions, and spontaneous abortions. Fertil
Steril 41,122–130.
Scher C, Haudenschild C and Klagsbrun M (1976) The chick chorioallantoic
membrane as a model system for the study of tissue invasion by viral
transformed cells. Cell 8,373–382.
Schor E, Baracat EC, Simoes MJ, de Freitas V, Giannotti Filho O and de Lima
GR (1999) Effects of conjugated estrogens and progestogen in surgically
induced endometriosis in oophorectomized rats. Clin Exp Obstet Gynecol
26,158–161.
Sharpe KL, Bertero MC, Muse KN and Vernon MW (1991) Spontaneous
and steroid-induced recurrence of endometriosis after suppression by a
gonadotropin-releasing hormone antagonist in the rat. Am J Obstet Gynecol
164,187–194.
Somigliana E, Vigano P, Rossi G, Carinelli S, Vignali M and Panina-
Bordignon P (1999) Endometrial ability to implant in ectopic sites can be
prevented by interleukin-12 in a murine model of endometriosis. Hum
Reprod 14,2944–2950.
Steinleitner A, Lambert H and Roy S (1991a) Immunomodulation with pen-
toxifylline abrogates macrophage-mediated infertility in an in vivo model:
a paradigm for a novel approach to the treatment of endometriosis-associated
subfertility. Fertil Steril 55,26–31.
Steinleitner A, Lambert H, Suarez M, Serpa N, Robin B and Cantor B (1991b)
Periovulatory calcium channel blockade enhances reproductive perform-
ance in an animal model for endometriosis-associated subfertility. Am J
Obstet Gynecol 164,949–952.
Story L and Kennedy S (2004) Animal studies in endometriosis: a review.
ILAR J 45,132–138.
Tabibzadeh S, Miller S, Dodson WC and Satyaswaroop PG (1999) An
experimental model for the endometriosis in athymic mice. Front Biosci
4,C4–C9.
Te Linde RW and Scott RB (1950) Experimental endometriosis. Am J Obstet
Gynecol 60,1147–1173.
Uchiide I, Ihara T and Sugamata M (2002) Pathological evaluation of the rat
endometriosis model. Fertil Steril 78,782–786.
Van Langendonckt A, Casanas-Roux F, Eggermont J and Donnez J (2004)
Characterization of iron deposition in endometriotic lesions induced in the
nude mouse model. Hum Reprod 19,1265–1271.
Vanden Heuvel JP, Clark GC, Tritscher A and Lucier GW (1994) Accumula-
tion of polychlorinated dibenzo-p-dioxins and dibenzofurans in liver of
control laboratory rats. Fundam Appl Toxicol 23,465–469.
Vercellini P, Trecca D, Oldani S, Fracchiolla NS, Neri A and Crosignani PG
(1994) Analysis of p53 and ras gene mutations in endometriosis. Gynecol
Obstet Invest 38,70–71.
Vernon MW and Wilson EA (1985) Studies on the surgical induction of
endometriosis in the rat. Fertil Steril 44,684–694.
Wheeler JM (1989) Epidemiology of endometriosis-associated infertility. J
Reprod Med 34,41–46.
Wolber EM, Kressin P, Meyhofer-Malik A, Diedrich K and Malik E (2003)
Differential induction of matrix metalloproteinase 1 and 2 in ectopic
endometrium. Reprod Biomed Online 6,238–243.
Wright JA and Sharpe-Timms KL (1995) Gonadotropin-releasing hormone
agonist therapy reduces postoperative adhesion formation and reformation
after adhesiolysis in rat models for adhesion formation and endometriosis.
Fertil Steril 63,1094–1100.
Yang JZ, Agarwal SK and Foster WG (2000) Subchronic exposure to 2,3,7,8-
tetrachlorodibenzo-p-dioxin modulates the pathophysiology of endometri-
osis in the cynomolgus monkey. Toxicol Sci 56,374–381.
Yano S, Ikegami Y and Nakao K (1996) Studies on the effect of the new non-
steroidal aromatase inhibitor fadrozole hydrochloride in an endometriosis
model in rats. Arzneimittelforschung 46,192–195.
Yao Z, Shen X, Capodanno I, Donnelly M, Fenyk-Melody J, Hausamann J,
Nunes C, Strauss J and Vakerich K (2005) Validation of rat endometriosis
model by using raloxifene as a positive control for the evaluation of novel
SERM compounds. J Invest Surg 18,177–183.
Zaino RJ, Satyaswaroop PG and Mortel R (1985) Histologic response of nor-
mal human endometrium to steroid hormones in athymic mice. Hum
Pathol 16,867–872.
Zamah NM, Dodson MG, Stephens LC, Buttram VC Jr, Besch PK and
Kaufman RH (1984) Transplantation of normal and ectopic human
endometrial tissue into athymic nude mice. Am J Obstet Gynecol
149,591–597.
Zeitoun KM and Bulun SE (1999) Aromatase: a key molecule in the pathophys-
iology of endometriosis and a therapeutic target. Fertil Steril 72,961–969.
Zeitoun K, Takayama K, Sasano H, Suzuki T, Moghrabi N, Andersson S,
Johns A, Meng L, Putman M, Carr B et al. (1998) Deficient 17beta-
hydroxysteroid dehydrogenase type 2 expression in endometriosis: failure
to metabolize 17beta-estradiol. J Clin Endocrinol Metab 83,4474–4480.
Zhou HE and Nothnick WB (2005) The relevancy of the matrix metalloprotei-
nase system to the pathophysiology of endometriosis. Front Biosci
10,569–575.
Zondervan KT, Weeks DE, Colman R, Cardon LR, Hadfield R, Schleffler J,
Trainor AG, Coe CL, Kemnitz JW and Kennedy SH (2004) Familial
aggregation of endometriosis in a large pedigree of rhesus macaques.
Hum Reprod 19,448–455.
Submitted on March 15, 2006; resubmitted on May 5, 2006; accepted on May 20,
2006
Downloaded
from
https://academic.oup.com/humupd/article/12/5/641/779916
by
guest
on
28
January
2022

More Related Content

Similar to Animal models in endometriosis research.pdf

Recent trends in the mnagement of fibriod
Recent trends in the mnagement of  fibriodRecent trends in the mnagement of  fibriod
Recent trends in the mnagement of fibriod
drmcbansal
 
Dissecting the Reduced Ovarian Reserve Seen with Ovarian Enometriomas with Th...
Dissecting the Reduced Ovarian Reserve Seen with Ovarian Enometriomas with Th...Dissecting the Reduced Ovarian Reserve Seen with Ovarian Enometriomas with Th...
Dissecting the Reduced Ovarian Reserve Seen with Ovarian Enometriomas with Th...
Crimsonpublisherssmoaj
 
The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...
Journal of Research in Biology
 
The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...
Journal of Research in Biology
 
Embryo implantation 7
Embryo    implantation 7Embryo    implantation 7
Embryo implantation 7
鋒博 蔡
 
Profiling the gene signature
Profiling the gene signatureProfiling the gene signature
Profiling the gene signature
t7260678
 

Similar to Animal models in endometriosis research.pdf (20)

International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI) International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)
 
Paget's Disease of the Breast
Paget's Disease of the BreastPaget's Disease of the Breast
Paget's Disease of the Breast
 
Diagnostic Biomarker Candidates Including NT5DC2 for Human Uterine Mesenchyma...
Diagnostic Biomarker Candidates Including NT5DC2 for Human Uterine Mesenchyma...Diagnostic Biomarker Candidates Including NT5DC2 for Human Uterine Mesenchyma...
Diagnostic Biomarker Candidates Including NT5DC2 for Human Uterine Mesenchyma...
 
Recent trends in the mnagement of fibriod
Recent trends in the mnagement of  fibriodRecent trends in the mnagement of  fibriod
Recent trends in the mnagement of fibriod
 
Dissecting the Reduced Ovarian Reserve Seen with Ovarian Enometriomas with Th...
Dissecting the Reduced Ovarian Reserve Seen with Ovarian Enometriomas with Th...Dissecting the Reduced Ovarian Reserve Seen with Ovarian Enometriomas with Th...
Dissecting the Reduced Ovarian Reserve Seen with Ovarian Enometriomas with Th...
 
Functional Assessment of Quality of Life Using EORTC QLQ-CR29 in Patients wit...
Functional Assessment of Quality of Life Using EORTC QLQ-CR29 in Patients wit...Functional Assessment of Quality of Life Using EORTC QLQ-CR29 in Patients wit...
Functional Assessment of Quality of Life Using EORTC QLQ-CR29 in Patients wit...
 
International Journal of Reproductive Medicine & Gynecology
International Journal of Reproductive Medicine & GynecologyInternational Journal of Reproductive Medicine & Gynecology
International Journal of Reproductive Medicine & Gynecology
 
Endometriosis - All most everything you need to know
Endometriosis - All most everything you need to knowEndometriosis - All most everything you need to know
Endometriosis - All most everything you need to know
 
Endometrium part 1 2018
Endometrium part 1 2018Endometrium part 1 2018
Endometrium part 1 2018
 
The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...
 
The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...The effect of microgravity on cell death, cell growth and cell cycle on breas...
The effect of microgravity on cell death, cell growth and cell cycle on breas...
 
Cytogenetic an Experimental Monitoring Test for Plant Extracts
Cytogenetic an Experimental Monitoring Test for Plant ExtractsCytogenetic an Experimental Monitoring Test for Plant Extracts
Cytogenetic an Experimental Monitoring Test for Plant Extracts
 
What is new in IVF(In Vitro Fertility)?
What is new in IVF(In Vitro Fertility)?What is new in IVF(In Vitro Fertility)?
What is new in IVF(In Vitro Fertility)?
 
Embryo implantation 7
Embryo    implantation 7Embryo    implantation 7
Embryo implantation 7
 
Mechanical signals inhibit growth of a grafted tumor in vivo proof of concept
Mechanical signals inhibit growth of a grafted tumor in vivo  proof of conceptMechanical signals inhibit growth of a grafted tumor in vivo  proof of concept
Mechanical signals inhibit growth of a grafted tumor in vivo proof of concept
 
Cervical Cystic Hygroma-a Case Report
Cervical Cystic Hygroma-a Case ReportCervical Cystic Hygroma-a Case Report
Cervical Cystic Hygroma-a Case Report
 
Profiling the gene signature
Profiling the gene signatureProfiling the gene signature
Profiling the gene signature
 
A case report of squamous cell carcinoma in buffalo.pdf
A case report of squamous cell carcinoma in buffalo.pdfA case report of squamous cell carcinoma in buffalo.pdf
A case report of squamous cell carcinoma in buffalo.pdf
 
Animal models
Animal modelsAnimal models
Animal models
 
Ovarian cancer
Ovarian cancerOvarian cancer
Ovarian cancer
 

Recently uploaded

🌹Attapur⬅️ Vip Call Girls Hyderabad 📱9352852248 Book Well Trand Call Girls In...
🌹Attapur⬅️ Vip Call Girls Hyderabad 📱9352852248 Book Well Trand Call Girls In...🌹Attapur⬅️ Vip Call Girls Hyderabad 📱9352852248 Book Well Trand Call Girls In...
🌹Attapur⬅️ Vip Call Girls Hyderabad 📱9352852248 Book Well Trand Call Girls In...
Call Girls In Delhi Whatsup 9873940964 Enjoy Unlimited Pleasure
 
Call Girls in Gagan Vihar (delhi) call me [🔝 9953056974 🔝] escort service 24X7
Call Girls in Gagan Vihar (delhi) call me [🔝  9953056974 🔝] escort service 24X7Call Girls in Gagan Vihar (delhi) call me [🔝  9953056974 🔝] escort service 24X7
Call Girls in Gagan Vihar (delhi) call me [🔝 9953056974 🔝] escort service 24X7
9953056974 Low Rate Call Girls In Saket, Delhi NCR
 
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls * UPA...
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls  * UPA...Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls  * UPA...
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls * UPA...
mahaiklolahd
 

Recently uploaded (20)

Trichy Call Girls Book Now 9630942363 Top Class Trichy Escort Service Available
Trichy Call Girls Book Now 9630942363 Top Class Trichy Escort Service AvailableTrichy Call Girls Book Now 9630942363 Top Class Trichy Escort Service Available
Trichy Call Girls Book Now 9630942363 Top Class Trichy Escort Service Available
 
Model Call Girls In Chennai WhatsApp Booking 7427069034 call girl service 24 ...
Model Call Girls In Chennai WhatsApp Booking 7427069034 call girl service 24 ...Model Call Girls In Chennai WhatsApp Booking 7427069034 call girl service 24 ...
Model Call Girls In Chennai WhatsApp Booking 7427069034 call girl service 24 ...
 
Independent Call Girls In Jaipur { 8445551418 } ✔ ANIKA MEHTA ✔ Get High Prof...
Independent Call Girls In Jaipur { 8445551418 } ✔ ANIKA MEHTA ✔ Get High Prof...Independent Call Girls In Jaipur { 8445551418 } ✔ ANIKA MEHTA ✔ Get High Prof...
Independent Call Girls In Jaipur { 8445551418 } ✔ ANIKA MEHTA ✔ Get High Prof...
 
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
 
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
 
🌹Attapur⬅️ Vip Call Girls Hyderabad 📱9352852248 Book Well Trand Call Girls In...
🌹Attapur⬅️ Vip Call Girls Hyderabad 📱9352852248 Book Well Trand Call Girls In...🌹Attapur⬅️ Vip Call Girls Hyderabad 📱9352852248 Book Well Trand Call Girls In...
🌹Attapur⬅️ Vip Call Girls Hyderabad 📱9352852248 Book Well Trand Call Girls In...
 
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
 
Call Girls in Delhi Triveni Complex Escort Service(🔝))/WhatsApp 97111⇛47426
Call Girls in Delhi Triveni Complex Escort Service(🔝))/WhatsApp 97111⇛47426Call Girls in Delhi Triveni Complex Escort Service(🔝))/WhatsApp 97111⇛47426
Call Girls in Delhi Triveni Complex Escort Service(🔝))/WhatsApp 97111⇛47426
 
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
 
Jogeshwari ! Call Girls Service Mumbai - 450+ Call Girl Cash Payment 90042684...
Jogeshwari ! Call Girls Service Mumbai - 450+ Call Girl Cash Payment 90042684...Jogeshwari ! Call Girls Service Mumbai - 450+ Call Girl Cash Payment 90042684...
Jogeshwari ! Call Girls Service Mumbai - 450+ Call Girl Cash Payment 90042684...
 
Call Girls in Gagan Vihar (delhi) call me [🔝 9953056974 🔝] escort service 24X7
Call Girls in Gagan Vihar (delhi) call me [🔝  9953056974 🔝] escort service 24X7Call Girls in Gagan Vihar (delhi) call me [🔝  9953056974 🔝] escort service 24X7
Call Girls in Gagan Vihar (delhi) call me [🔝 9953056974 🔝] escort service 24X7
 
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
 
Call Girls Rishikesh Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Rishikesh Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Rishikesh Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Rishikesh Just Call 8250077686 Top Class Call Girl Service Available
 
VIP Service Call Girls Sindhi Colony 📳 7877925207 For 18+ VIP Call Girl At Th...
VIP Service Call Girls Sindhi Colony 📳 7877925207 For 18+ VIP Call Girl At Th...VIP Service Call Girls Sindhi Colony 📳 7877925207 For 18+ VIP Call Girl At Th...
VIP Service Call Girls Sindhi Colony 📳 7877925207 For 18+ VIP Call Girl At Th...
 
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls * UPA...
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls  * UPA...Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls  * UPA...
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls * UPA...
 
Call Girls Visakhapatnam Just Call 8250077686 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 8250077686 Top Class Call Girl Service Ava...Call Girls Visakhapatnam Just Call 8250077686 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 8250077686 Top Class Call Girl Service Ava...
 
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 9332606886 𖠋 Will You Mis...
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 9332606886 𖠋 Will You Mis...The Most Attractive Hyderabad Call Girls Kothapet 𖠋 9332606886 𖠋 Will You Mis...
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 9332606886 𖠋 Will You Mis...
 
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
 
Top Rated Hyderabad Call Girls Chintal ⟟ 9332606886 ⟟ Call Me For Genuine Se...
Top Rated  Hyderabad Call Girls Chintal ⟟ 9332606886 ⟟ Call Me For Genuine Se...Top Rated  Hyderabad Call Girls Chintal ⟟ 9332606886 ⟟ Call Me For Genuine Se...
Top Rated Hyderabad Call Girls Chintal ⟟ 9332606886 ⟟ Call Me For Genuine Se...
 
Call Girls Kakinada Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kakinada Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kakinada Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kakinada Just Call 9907093804 Top Class Call Girl Service Available
 

Animal models in endometriosis research.pdf

  • 1. © The Author 2006. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology. All rights reserved. For Permissions, please email: journals.permissions@oxfordjournals.org 641 Human Reproduction Update, Vol.12, No.5 pp. 641–649, 2006 doi:10.1093/humupd/dml026 Advance Access publication June 14, 2006 Animal models in endometriosis research Ruth Grümmer Institute of Anatomy, University Hospital Essen, Essen, Germany E-mail: ruth.gruemmer@uni-essen.de Endometriosis is a common gynaecological disease, defined as the presence of endometrial tissue outside the uterus, causing pelvic pain and subfertility in ~10% of women of reproductive age. Current therapies lead to pain relief, however, do not address the causes and entail severe side effects. Still little is known about the pathogenic processes leading to the development and maintenance of endometriosis. Because endometriosis occurs spontaneously only in humans and some non-human primates, animal models of induced endometriosis have been developed and are of high value for the evaluation of pathophysiological mechanisms underlying the development of this disease. These experimental models include the autotransplantation of uterine fragments into the peritoneal cavity of rodents and non-human primates or the heterotransplantation of human endometrial or endometriotic tissue to immunodeficient mice or onto the chicken chorioallantoic membrane (CAM). This review describes the animal models for endometrio- sis and assesses their different potentials and limitations in regard to endometriosis research, with the aim of develop- ing novel non-invasive diagnostic tools and improved strategies for the treatment of endometriosis in women. Key words: animal model/CAM/endometriosis/non-human primates/rodents The need for animal models in endometriosis research Endometriosis is one of the most frequent benign gynaecological diseases; it is thought to occur in 7–10% of women (Wheeler, 1989) but may even affect up to 60% of women of reproductive age with pelvic symptoms or disturbance of fertility (Pellicer et al., 2001; Giudice and Kao, 2004). Yet, little is known about the pathogenic processes leading to the development and maintenance of this disease, and currently available medical therapies are unsat- isfactory, because they focus on treating the symptoms rather than curing the causes. In addition, they cannot be used for prolonged duration because of severe secondary side effects (Rice, 2002). Thus, there is a definite need to develop new drugs to provide spe- cific and more efficient therapeutic alternatives that eliminate endometriotic lesions, prevent recurrences and do not interfere with the fertility potential. One widely accepted mechanism for the development of perito- neal endometriotic lesions is the adhesion and growth of endome- trial fragments deposited into the peritoneal cavity via retrograde menstruation (Sampson, 1927). However, because retrograde menstruation occurs in nearly all women of reproductive age (Halme et al., 1984), impairment of additional factors is needed for the establishment and maintenance of these ectopic lesions. Because, at the time of clinical presentation, most women already have established endometriosis, it is hardly possible to give exper- imental evidence for physiological roles in the pathogenesis of this disease in humans. In addition, ethical considerations limit the performance of controlled experiments, and it is not possible to monitor the disease progression without performing repeated laparoscopies. Thus, research into the fundamental mechanisms by which menstrual endometrium adheres, invades and establishes a functional vasculature to persist in an ectopic site, as well as the development of new therapeutical approaches, is best performed in experimental animal models. Because menstrual shedding is a requirement for the spontane- ous development of this disease, endometriosis occurs naturally only in humans and some non-human primates. Non-human pri- mates have been extensively used for the investigation of endome- triosis; however, the very high costs of animal handling limit the use of monkeys as an experimental model. For this reason, the establishment of small laboratory animals, especially rodents, as models for endometriosis by the transplantation of pieces of endometrial tissue to ectopic sites has greatly extended in recent years. This review provides a survey of commonly used animal models and their application for different challenges. CAM assay Characterization of the CAM assay The chicken chorioallantoic membrane (CAM) model can be con- sidered as an animal model in the broader sense. Originally, it had been developed to study the invasive, metastatic and angiogenic potential of neoplastic cells (Scher et al., 1976; Armstrong et al., 1982). It has been established as a model for endometriosis by cul- turing fragments of human endometrial tissue on the basal layer of the CAM of fertilized chicken eggs after prior incubation for 7–10 days (Malik et al., 2000; Maas et al., 2001). Endometrial fragments Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022
  • 2. R.Grümmer 642 from the proliferative and secretory phase of the menstrual cycle as well as the menstrual endometrium invade across the epithe- lium into the mesenchymal layer and develop endometriosis-like lesions in this layer of the CAM within 3 days after grafting of the human tissue (Maas et al., 2001; Nap et al., 2003, 2004). It was shown that these endometrial fragments needed to contain intact glandular structures as well as stromal components. Application of the CAM assay Angiogenesis could be documented by ingrowth of vessels from the CAM into the human endometrial fragments (Maas et al., 2001). The anti-angiogenic factors endostatin, TNP-470 and anti-vascular endothelial growth factor (VEGF) antibodies all significantly decreased the angiogenic response to endometrial transplants (Nap et al., 2005). Endometrial fragments of women with and without endometriosis showed no significant differences in VEGF expression or angiogenic induction in this assay (Gescher et al., 2005). In contrast, Oosterlynck et al. (1993) demonstrated that peritoneal fluid of women with endometriosis induced significantly higher angiogenic responses in the CAM than peritoneal fluid of women without endometriosis. Because the CAM contains extracellular matrix components similar to the ECM of the human peritoneum (Giannopoulou et al., 2001), it can be used to investigate the invasive potential of endometriotic tissue. Nap et al. (2004) demonstrated that the expression profiles of matrix metalloproteinases (MMPs) were similar in experimentally induced endometriosis in CAMs compared with human endometriosis and that the development of early endometriotic lesions in the CAM model was pre- vented by inhibiting MMP-1, -2, -3, -7 and -13 activity. According to Wolber et al. (2003), relative concentrations of MMP-1 mRNA, but not MMP-2 mRNA, increased strongly after culture on the CAM. Evaluation of the CAM assay The CAM assay is well suited for the investigation of mechanisms involved in invasion and angiogenesis during the first steps in the establishment of endometriotic lesions, for example, by the inhibi- tion of factors known to participate in these processes. The ectopic lesions are easily accessible and can be simply monitored during the course of an experiment. This endometriosis model is hardly applicable for the investigation of immunological or inflammatory responses or to analyse long-term drug effects in the course of the disease. Rodent models In contrast to humans and non-human primates, estrous animals do not shed their endometrial tissue and therefore do not develop endometriosis spontaneously. However, endometriosis can be induced by transplanting endometrial tissue to ectopic sites. These models are classified into two types, homologous and heterologous models. Homologous models have been employed utilizing the sur- gical transplantation of endometrium of the same or syngeneic ani- mals in immunocompetent animals, whereas in heterologous models, human endometrial fragments are transferred either intra- peritoneally or subcutaneously to immunodeficient mice. Autotransplantation of endometrium to immunocompetent mice and rats Characterization of the homologous model Autotransplantation of uterine tissue to ectopic sites of small labo- ratory animals has been established not only in rats (Golan et al., 1984; Jones, 1984; Vernon and Wilson, 1985; Rajkumar et al., 1990; Sharpe et al., 1991) and mice (Cummings and Metcalf, 1995; Somigliana et al., 1999; Rossi et al., 2000) but also in ham- sters (Steinleitner et al., 1991b) and rabbits (Schenken and Asch, 1980; Dunselman et al., 1989; Homm et al., 1989; Rock et al., 1993). Of those non-primate models, mainly rat and mouse mod- els have been further developed in recent years. In these models of endometriosis, uteri are removed and cut into small pieces which are then reintroduced, mostly by suturing, into the peritoneal cav- ity. In most of these studies, endometrium was not separated from myometrium; thus, both compartments were implanted (Vernon and Wilson, 1985; Cummings and Metcalf, 1995). In the rat, the uterine tissue develops into fluid-filled, ovoid, cystic structures composed of myometrial and endometrial tissue. The cysts grow but stabilize their size by ∼2 months and remain viable for at least 10 months (Vernon and Wilson, 1985). In the mouse, the ectopic uterine fragments show histological characteristics of the human disease, including the formation of multiple, highly vascularized lesions containing endometrial glands, stroma and cysts, independent of their peritoneal localiza- tion in the abdomen (Cummings and Metcalf, 1995). A limited number of experiments separating the endometrium from the myo- metrium, only inoculating the endometrium to ectopic sites, have been performed in rats (Katsuki et al., 1998) and mice (Somigliana et al., 1999; Hirata et al., 2005; Yao et al., 2005). In the study of Somigliana et al. (1999), isolated endometrial tissue was minced and reintroduced into the peritoneal cavity of recipi- ent, syngeneic animals. Both donor and recipient mice had been ovariectomized and received estrogen treatment. In this study, all recipient animals revealed evidence of peritoneal endometriosis after 3 weeks, and neovascularization was observed at the surface of the lesions. However, the ‘take-rate’, that is the number of lesions recovered after a known number of endometrial fragments have been randomly inoculated, was on average 30% of the inocu- lated tissue. For better identification of size and localization of ectopic endometriotic lesions after transplantation, Hirata et al. (2005) developed a homologous mouse model using ‘green mice’ that ubiquitously express green fluorescent protein (GFP) as donors and transplanted the fluorescent endometrial tissue into recipient mice. They could show that the weight of endometriotic lesions significantly correlated with the measured fluorescence intensity. This fluorescence was significantly higher in the estrogen- supplemented mice as compared with control animals, supporting the estrogen dependency of these ectopic endometrial lesions. Application of the homologous model Endometriosis is known to be an estrogen-dependent disease in women, and the suppression of the serum estrogen level supports the regression of those ectopic lesions (Lapp, 2000; Rice, 2002). Autotransplanted uterine tissue in the rodent model shows steroid hormone dependency, and thus, this model has been widely used for determining the responsiveness of ectopic lesions to steroid Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022
  • 3. Animal models in endometriosis research 643 hormones as well as to drugs interfering with steroid action. Cor- responding to the situation in humans, the growth of the ectopic endometrial tissue in both rodent species was shown to be estro- gen dependent (Vernon and Wilson, 1985; Rajkumar et al., 1990; Rossi et al., 2000). In rats which had been ovariectomized 3 weeks after engrafts of uterine tissue, the ectopic fragments recovered much better in animals treated with estrogen alone after ovariec- tomy than in those with combined estrogen and progesterone treat- ment (Schor et al., 1999). Fang et al. (2004) confirmed the important role for estrogen as a factor determining the size of the implants in mice and demonstrated that the antiproliferative effect of progesterone on the estrogen-dependent growth of endometriotic tissue is mediated by intact progesterone receptor, because this estrogen-dependent growth could be suppressed by progesterone in uterine tissues of wild-type but not of progester- one receptor-deficient mice. According to the situation in women, the regression of uterine ectopic implants could be induced in rats by generating a hypoes- trogenic state, for example, by ovariectomy (Kudoh et al., 1997), by the application of GnRH agonists (Sakata et al., 1990; Wright and Sharpe-Timms, 1995; Kudoh et al., 1997), by synthetic pro- gestational compounds such as levonorgestrel or dienogest (Jones, 1984; Katsuki et al., 1998) or by danazol treatment (Sakata et al., 1990). Corresponding effects could be achieved in this model by reducing the estrogen concentration by the application of anti- estrogens (Saito et al., 2003), by using the selective estrogen receptor modulator raloxifen (Yao et al., 2005) or by treatment with aromatase inhibitors which interfere with estrogen synthesis (Yano et al., 1996; Kudoh et al., 1997). Moreover, the autologous rat model in particular has been extensively used for studies of immune-modulating drugs and anti-inflammatory agents in endometriosis. Uchiide et al. (2002) demonstrated that uterine autotransplantation in rats induces the infiltration of allergic inflammatory-related cells in peritoneal stroma attached to the ectopic uterine tissue. Immunomodulation by intraperitoneal or subcutaneous treatment with interferon-α-2b in the rat reduced the size of induced lesions as monitored by con- secutive laparotomies over a period of up to 4 months (Ingelmo et al., 1999) as did the application of the immunomodulator loxoribine in rats (Keenan et al., 1999) and the intraperitoneal injection of interleukin-12 in a syngeneic mouse model (Somigliana et al., 1999). In addition, the development of experimentally induced endometriosis in rats was inhibited by recombinant human tumour necrosis factor (TNF)-binding protein-1 (r-hTBP-1), which is the soluble form of TNF receptor type I (D’Antonio et al., 2000). This effect was also seen after treatments of rats with pentoxifylline, an anti-inflammatory substance reported to reduce the production of inflammatory cytokines without induc- ing a hypoestrogenic state (Nothnick et al., 1994), as well as with ciglitazone, a ligand for proliferator-activated receptor-γ (PPAR-γ) (Lebovic et al., 2004). Moreover, the initial develop- ment of ectopic implants in rats has been reduced by cyclooxy- genase-2 (COX-2) inhibitors (Matsuzaki et al., 2004) and also by the application of various non-steroidal anti-inflammatory drugs such as celecobix, indomethacin, sulinac and ibuprofen (but not aspirin) in a mouse model for endometriosis (Efstathiou et al., 2005). Furthermore, this rodent model provides the opportunity to investigate the effect of environmental contaminants on the estab- lishment of ectopic uterine implants. It has been demonstrated that exposure to dioxin preceding the surgical induction of endometri- osis produces a dose-dependent increase in endometriotic site diameter in rats and mice, which was much stronger in mice (Cummings et al., 1996). Recently, in an interesting approach, Dinulescu et al. (2005) described the first mouse model of de-novo endometriosis. Activa- tion of the oncogene K-ras in ovarian surface epithelial cells, but not in cells of the peritoneal lining, resulted in the development of benign epithelial lesions with histomorphological features of human endometriosis. In addition to developing ovarian lesions, nearly half the mice also developed peritoneal endometriosis 8 months after the K-ras activation of ovarian surface epithelial cells. In combination with a conditional deletion of Pten, which has been implicated in the progression of ovarian endometriosis to endometrioid ovarian carcinoma in humans (Sato et al., 2000), K- ras expression even induced metastatic endometrioid ovarian ade- nocarcinomas. However, to date, no mutations of K-ras have been found in human endometriosis (Vercellini et al., 1994; Otsuka et al., 2004). The hallmark symptoms of endometriosis in women include severe pelvic pain and significantly reduced fertility (Elsheikh et al., 2003). The influence of ectopic lesions on fertility has been investigated in laboratory rodents. Although Cummings and Metcalf (1996) did not observe a decrease in fertility in mice, a reduced fecundity could be observed in rats with artificially induced endometriosis (Vernon and Wilson, 1985), which could be due in part to an increased number of luteinized unruptured ovarian follicles (Moon et al., 1993). In addition, a role of pelvic adhesions has not be excluded. Moreover, peritoneal inflamma- tory cell hyperactivation could have an effect on fertility in endometriosis. This was supported by the work of Steinleitner et al. (1991a), demonstrating that macrophage-mediated subfertil- ity in the mouse model could be compensated by pentoxifylline, a drug that reverses the effect of macrophage hyperactivation, and that the drug-mediated inhibition of macrophage activation enhanced fertility in a hamster endometriosis model (Steinleitner et al., 1991b). Recently, studies have been published investigating the effect of ectopic endometrial lesions on pain behaviour in the rat model of endometriosis. Here, the autotransplanted ectopic endometrial cystic fragments develop their own innervation including sympa- thetic efferent as well as sensory fibres (Berkley et al., 2004), which may have a general influence on the nervous system. This is supported by the finding that vaginal nociception was increased in rats with endometrial cysts similar to the situation in women with endometriosis. In addition, those rats also displayed vaginal hyperalgesia (Berkley et al., 2001), one of the symptoms of increased pelvic pain in women. It is speculated that neuroactive agents contained in the endometrial cysts may activate nociceptive afferents influencing central neural mechanisms associated with vaginal nociception as well as with reproduction via viscero– visceral interactions (Cason et al., 2003; Berkley et al., 2004). It remains to be elucidated whether the rats exhibit chronic pelvic pain symptoms other than vaginal hyperalgesia in this model of endometriosis. Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022
  • 4. R.Grümmer 644 Evaluation of the homologous model There are relevant differences between rodent and human repro- ductive physiology; consequently, these models possess limita- tions. Because rodents do not menstruate, they do not develop spontaneous endometriosis, and the disease has to be induced artificially by the autotransplantation of uterine tissue. In addi- tion, in most of these rodent studies, the uterine fragments which were transplanted included the myometrial layer which could affect the development of the lesions. When evaluating the weight and size of the ectopic lesions, the myometrium could make up a considerable proportion, not representing the situation of human endometriotic lesions. Despite these limitations, the rodent model offers significant advantages. Apart from the limited costs and the opportunity to perform studies in large groups of genetically similar animals, long-term studies can be performed because there is no rejection of the autotransplanted ectopic tissue. It is well suited for the investigation of mechanisms involved in the peritoneal attachment of endometrial cells as well as of effects of therapeutical drugs and chemicals, and it allows the evaluation of ectopic lesions at differ- ent time intervals. Moreover, the homologous rodent model of sur- gically induced endometriosis provides a well-characterized immune system and thus is appropriate and extensively used to study the effect of immune-modulating drugs and anti-inflamma- tory agents on endometriosis. Furthermore, in regard to new insights in the aetiology of endometriosis obtained by genomic and proteomic approaches, transgenic mice models may gain importance as models of endometriosis to examine functions driven by single genes. Xenotransplantation of human endometrium to immunodeficient mice Characterization of the heterologous model Because causal factors for the development and maintenance of endometriosis could originate in the endometrium itself and may not be present in the rodent endometrium, models have been developed by implanting human endometrial tissue instead of autologous endometrium into immunodeficient mouse strains. Most widely used for this purpose is the athymic nude mouse, which lacks mature T lymphocytes. Human endometrial fragments from the proliferative or secretory phase of the menstrual cycle (Zamah et al., 1984; Bergqvist et al., 1985; Zaino et al., 1985; Bruner et al., 1997; Nisolle et al., 2000; Grummer et al., 2001; Beliard et al., 2002) as well as the menstrual endometrium (Nisolle et al., 2000; Van Langendonckt et al., 2004; Eggermont et al., 2005) have been successfully implanted either subcutane- ously or intraperitoneally into nude mice. These fragments implant and form endometriotic-like lesions which resemble those found in patients in terms of macroscopic and histological appearance, whereas the stage of the menstrual cycle at the time of collecting the human tissue seems to have no impact on the development of those ectopic lesions (Nisolle et al., 2000). Preservation of estro- gen and progesterone receptors and steroid responsiveness have been demonstrated in the ectopic human tissue (Zamah et al., 1984; Bruner et al., 1997, 1999; Grummer et al., 2001). Angio- genesis guarantees the maintenance of the transplants and the transport of systemically applied drugs to the human endometrial tissue. The establishment of blood vessels of mouse origin occurred from just 4 days after transplantation onwards, independent of the localization of the ectopic lesions (Grummer et al., 2001; Hull et al., 2003). When endometrial tissue was randomly inoculated into the peri- toneal cavity, peritoneal adhesion occurred from 2 days after implantation onwards. The sites of implantation of the endometrial fragments were preferentially the gut, the muscles of the abdominal wall, the liver and the adipose fat surrounding abdominal organs (Grummer et al., 2001; Fortin et al., 2003). However, studies from our group as well as others revealed that the recovery rate is highly variable from one animal to another and in average does not exceed 30% (Bruner et al., 1997; Grummer et al., 2001). This ‘take-rate’ is further reduced after in vitro culturing of endometrial tissue for 24 h before transplantation but could be enhanced to 100% by suturing the human endometrial fragments to different intraperitoneal sites (Grummer et al., 2001). This is favourable, in regard to drug test- ing, because a difference in the number of lesions in the two groups of animals could be due to different ‘take-rates’ at the beginning of the experiment rather than to the effect of a drug. For non-invasive detection and quantification of implanted endometrial tissue, Tabibzadeh et al. (1999) loaded dissociated glands and stromal cells with a lipophylic fluorescent dye (DiO) before in vivo transplantation. However, an effect of the solvent [dimethylsulphoxide (DMSO) and absolute ethanol] on the endometrial tissue cannot be excluded. An interesting approach that is better for monitoring of subcutaneously or intraperitoneally transplanted endometrial fragments was recently described by Fortin et al. (2003). They transduced endometrial fragments of the proliferative phase with GFP cDNA before implantation, resulting in a transient fluorescence of the endometrial tissue allowing opti- cal in vivo imaging of the implantation and development of ectopic lesions. Because adenoviruses are transient expression vectors, they observed that fluorescence of endometrial tissue fades out around the third or fourth week, limiting the non-inva- sive follow-up to this time frame. In addition, when using GFP as a reporter gene, in vivo quantification would probably be limited to subcutaneous lesions, which can be directly visualized through the skin of living mice, because the wavelength of GFP-emitted light from intraperitoneal fragments is absorbed by surrounding tissues. Thus, this model allows dynamic studies of lesion by mon- itoring implantation and development. However, a yet unknown interaction between GFP and lesions or applicated drugs might interfere with the studies. Because, in the heterologous mouse model, human tissue is transplanted, the time frame for maintenance of intact and well- preserved tissue is limited. In most of the studies performed, cul- ture of human endometrium in nude mice does not exceed a period of 4 weeks. From 3 weeks after inoculation onwards, dedifferenti- ation parameters and lymphocyte infiltration can be observed (Grummer et al., 2001). Because longer maintenance of human tissue in this animal model would be advantageous for special experimental approaches, mice with an even more defective immune system have been established as heterologous models for endometriosis. Human endometrial tissue has successfully been transplanted to severe combined immunodeficient (SCID) mice (Aoki et al., 1994; Kaufmann et al., 1995; Awwad et al., 1999) as well as to non-obese diabetic (NOD)-SCID mice (Grummer et al., 2001), Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022
  • 5. Animal models in endometriosis research 645 both of which show a combined congenital deficiency in T- and B- lymphocyte function. Compared with the nude mouse model, human endometrium engrafted in SCID/NOD-SCID mice revealed a higher ‘take-rate’ of fragments (Aoki et al., 1994) and a better preserved morphology and expression of steroid hormone receptors over a period of 4 weeks (Grummer et al., 2001). Although these mice lack B and T lymphocytes, they do have some natural killer (NK) cell activity. Recently, Matsuura-Sawada et al. (2005) cultured human endometrial tissue subcutaneously in NOD/SCID/γ Cnull (NOG) immunodeficient mice which are addi- tionally defective in NK-cell activity. In this model, they repro- duced one human menstrual cycle of 28 days by hormone application in ovariectomized animals. At the level of histology, they showed cyclic changes also observed in eutopic human endometrium but without signs of decidualization. Greenberg and Slayden (2004) subcutaneously engrafted human endometriotic tissue in transgenic recombinase-activating gene-2 knockout [RAG-2/γ(c)KO] mice, which are not only devoid of B and T lymphocytes but also lack NK cells. Only endometriotic biopsy specimens that contained endometrium-like glands developed visible lesions. Red peritoneal lesions, but not biopsy specimens of ovarian endometrioma, established grafts at a high rate. In this animal model for endometriosis, they mimicked four consecutive human menstrual cycles by applying estrogen and progesterone via hormone-releasing capsules. The established grafts were well-vascularized and maintained steroid hormone receptors. Application of the heterologous model Animal models of endometriosis based on the xenotransplantation of human endometrium permit the elucidation of mechanisms involved in the establishment of endometriosis as well as testing the effects of potential therapeutical compounds on human tissue. One approach to prevent the development of peritoneal endometri- osis could target the adherence of endometrial tissue to and the invasion of the peritoneal lining. MMPs are involved in such inva- sion processes (Hudelist et al., 2005; Zhou and Nothnick, 2005). It could be shown that MMPs are regulated by steroid hormones and cytokines in endometrial tissue and that the suppression of MMPs inhibits the establishment of ectopic lesions by human endometrium in nude mice (Bruner et al., 1997, 1999; Bruner- Tran et al., 2002). Another therapeutical target discussed is the development of new blood vessels, which is essential for the persistence of ectopic endometriotic lesions. Endometrial xenografts in nude mice have been shown to produce high levels of VEGF (Nisolle et al., 2000), and anti-angiogenic agents such as soluble VEGF receptor (sflt-1) and VEGF-A-antibodies significantly inhibited the growth of endometriotic lesions in nude mice (Hull et al., 2003). Because endometriosis is an estrogen-dependent disease, the responsiveness of human ectopic lesions to steroid hormones has been investigated in this mouse model. Pretreatment of human endometrial cells with estrogen alone or in combination with a progestin, but not with progestin alone, before injection into the peritoneal cavity of nude mice resulted in a higher percentage of animals developing endometriotic-like lesions (Beliard et al., 2002). Using optical in vivo imaging, Fortin et al. (2004) demon- strated a significantly reduced lesion size in estrogen-untreated versus estrogen-treated ovariectomized nude mice. Recent studies have identified aberrations in the expression of aromatase and 17β-hydroxysteroid reductase-2 in human endome- triotic lesions (Zeitoun et al., 1998; Bulun et al., 1999; Zeitoun and Bulun, 1999), suggesting a role for steroid-metabolizing enzymes in the pathogenesis of endometriosis. We could demon- strate that the transcription of estrogen-metabolizing enzymes is maintained in human endometrial tissue cultured in nude mice and is regulated by the exogenously applied drugs danazol, dydroges- terone and medroxyprogesterone acetate, which are used in the treatment of endometriosis and are known to interact with the estrogen metabolism, as well as by an aromatase inhibitor (Gruemmer et al., 2005). In contrast to the homologous rodent model, mice used in the heterologous models have a deficient immune system and thus are hardly used in studies evaluating the effect of immune-modulating drugs. Application of an inhibitor of COX-2, which is known to be up-regulated at sites of inflammation, had no influence on the number or size of ectopic endometrial lesions in nude mice (Hull et al., 2005). However, anti-inflammatory components, contained for example in peritoneal fluid of women, could be investigated in this model. Tabibzadeh et al. (1999) demonstrated that endome- trial cells suspended in peritoneal fluid of women without endometriosis established significantly less ectopic lesions than those suspended in peritoneal fluid of women with endometriosis after intraperitoneal injection into nude mice. Evaluation of the heterologous model This model shares the advantages described for the homologous model, such as easy availability and low costs, as well as some of the limitations such as differences between rodent and human physiology. However, it does not provide an intact immune sys- tem. Yet, the heterologous mouse model for endometriosis repre- sents a promising tool for experimental approaches evaluating the aetiology of this disease and for therapeutic testing of pharmacolog- ical and hormonal modulations because it is based on human endometrium. For these investigations, human endometrial tissue can be taken from women with and without endometriosis as well as from endometriotic lesions. Regulatory mechanisms, for example, after drug administration, can be evaluated in the human ectopic tis- sue in an in vivo situation. Long-term studies that are not feasible in women can be performed, and the hormonal status of the human menstrual cycle can be mimicked in ovariectomized animals. Recently established methods using fluorescence allow in vivo imaging and may improve the quantification of both size and number of the ectopic lesions. In addition, techniques using mag- netic resonance imaging (MRI) for in vivo monitoring of the lesions are in progress. Primate models Characterization of the primate model Menstruating primates develop spontaneous endometriosis, estab- lishing ectopic lesions histologically identical and at similar sites compared with the human disease (MacKenzie and Casey, 1975; D’Hooghe et al., 1991; Dick et al., 2003). Spontaneous endometri- osis does not develop with a high frequency in all monkeys but could be shown to increase with time in capture possibly because of less pregnancies leading to a higher number of consecutive Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022
  • 6. R.Grümmer 646 menstrual cycles with increased exposure to retrograde menstrua- tion (D’Hooghe et al., 1996a). Although spontaneous endometrio- sis has been reported for 11 species of non-human primates (for review, see Story and Kennedy, 2004), most studies have been performed in rhesus macaques and baboons. Because spontaneous endometriosis in monkeys develops slowly over a period of several years, methods have been employed for the artificial induction of this disease. In the first studies, menstrual flow was diverted into the abdomen by reposi- tioning the cervix of rhesus monkeys (Te Linde and Scott, 1950) and later by occluding the cervix to increase the amount of retro- grade menstruation in baboons (D’Hooghe et al., 1994). In addi- tion, surgical induction of the disease can be performed by suturing fragments of endometrial tissue at ectopic sites or by seeding the peritoneal cavity with fragments of endometrial tissue (D’Hooghe et al., 1995b; Yang et al., 2000; Fazleabas et al., 2002). D’Hooghe et al. (1995b) induced endometriosis in a baboon model by intraperitoneal injection of endometrial tissue, demonstrating that endometriosis developed similarly to that observed in the spontaneous disease and that ectopic lesions were induced more efficiently by inoculating menstrual endometrium compared with luteal phase endometrium. Although, in this study, menstrual endometrium was extracted from each baboon by uter- ine curettage, thus also containing endometrial cell layers not physiologically shed during menstruation, other studies used shed menstrual endometrium obtained using a pipelle (Fazleabas et al., 2002, 2003). By this means, induced ectopic endometrial lesions in the baboon could be shown to express estrogen receptor-β, MMP-7, VEGF and aromatase (Fazleabas et al., 2002, 2003). Ongoing efforts focus on the non-invasive diagnosis for the identifi- cation of intraperitoneal lesions using MRI (Zondervan et al., 2004). These methods of induced endometriosis in non-human pri- mates have been extensively reviewed previously (D’Hooghe, 1997; D’Hooghe and Debrock, 2002; Story and Kennedy, 2004; Fazleabas, 2005). Application of the primate model Many studies have been performed evaluating spontaneous endometriosis on the one hand and using artificially induced endometriosis on the other hand. It could be shown that elevated estrogen levels as well as fre- quent hysterotomies, but not laparoscopies, lead to an increased risk for the development of spontaneous endometriosis (Hadfield et al., 1997) and that genetic predisposition also seems to contrib- ute to a higher risk for this disease (Zondervan et al., 2004). Regression of peritoneal endometriotic lesions could be demon- strated after the application of progesterone receptor modulators, either alone or in combination with a GnRH analogue, in induced endometriosis in cynomologous monkeys (Grow et al., 1996). The influence of environmental compounds on the development of spontaneous endometriosis was proven by exposure of rhesus monkeys to dioxin or polychlorinated biphenyls (PCBs), which resulted in a dose-dependent increase in incidence and severity of endometriosis (Rier et al., 1993; Vanden Heuvel et al., 1994; DeVito et al., 1995). The primate model is well suited to investigate the role of the immune system on the development and progression of endome- triosis. Subclinical peritoneal inflammation occurs in baboons during menstruation as well as after the intrapelvic injection of endometrium, as demonstrated by an increase in peritoneal fluid leukocyte concentration and the amount of peritoneal fluid cells positive for TNF-α, transforming growth factor (TGF-β1) and cer- tain leukocyte markers (D’Hooghe et al., 2001). It was shown that immunosuppression led to an increase in the number and size of spontaneously developed endometriotic lesions (D’Hooghe et al., 1995a). Neutralization of TNF-α with r-hTBP-1 inhibits the development of endometriotic lesions and adhesions after the arti- ficial induction of the disease (D’Hooghe et al., 2006), and a decrease in active red lesion number and surface area was observed in female baboons with spontaneous peritoneal endome- triosis after neutralizing TNF activity by treatment with etanercept (Barrier et al., 2004). With regard to the impact of endometriosis on fertility, it was proven that mild-to-severe endometriosis leads to lower preg- nancy rates in baboons (D’Hooghe et al., 1996b) as well as to a reduction in chemical and term pregnancy rates in another monkey species, Macaca fascicularis (Schenken et al., 1984). Whether these effects on fertility are due to peritoneal adhesions and/or to immunological responses is still to be investigated. In addition, a feedback effect of the endometriotic lesions on the eutopic endometrium is discussed, and it has to be evaluated whether the differences in fecundity result in, or are the result of, endometrio- sis. Whether the peritoneal presence of ectopic implantation sites influences the physiology of the eutopic endometrium can effec- tively be investigated in the primate model. Recently, an increase in the angiogenic factor Cyr61, which is known to be deregulated in the endometrium of women with endometriosis (Absenger et al., 2004), could be demonstrated in the eutopic endometrium of baboons following peritoneal inoculation with menstrual endometrium (Gashaw et al., 2006). This study provides evidence for a feedback mechanism from established lesions to induce a shift in gene expression patterns in the eutopic endometrium in the baboon. Evaluation of the primate model There is no doubt that primate models for endometriosis are most closely related to the situation in humans and thus are most appro- priate to investigate the numerous aspects of this disease, includ- ing the initiation and progression of endometriosis, immunological parameters, inheritance patterns as well as feedback mechanisms on the eutopic endometrium. They reveal many similarities to women with respect to pelvic anatomy, reproductive physiological characteristics and immunological features. Primates develop endometriosis spontaneously, and the disease can also be induced for research purposes. They represent the only experimental model, allowing the comparison of parameters in spontaneous endometriosis with parameters before and after disease induction, thus gaining information about cause and effect of the disease. However, ethical considerations and particularly the high costs of performing experiments using primates limit the use of this model in endometriosis research. Conclusions Animal models of endometriosis are of extreme value and indis- pensable for the evaluation of pathophysiological mechanisms Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022
  • 7. Animal models in endometriosis research 647 underlying the development of this prevalent gynaecological dis- ease. This review summarizes different commonly used animal models, their potentials limitations. These have to be weighted up when designing experimental approaches with regard to their applicability, for example, considering species-specific character- istics of metabolism or reproductive physiology, as well as to their cost–effect ratio. New insights into the aetiology of endometriosis will be obtained using genomic and proteomic approaches. In this regard, transgenic mice models might gain importance as models of endometriosis to examine functions driven by single genes. In gen- eral, animal models will help to develop novel non-invasive diag- nostic tools and improved therapeutical approaches for a better treatment of endometriosis in women. References Absenger Y, Hess-Stumpp H, Kreft B, Kratzschmar J, Haendler B, Schutze N, Regidor PA and Winterhager E (2004) Cyr61, a deregulated gene in endometriosis. Mol Hum Reprod 10,399–407. Aoki D, Katsuki Y, Shimizu A, Kakinuma C and Nozawa S (1994) Successful heterotransplantation of human endometrium in SCID mice. Obstet Gyne- col 83,220–228. Armstrong PB, Quigley JP and Sidebottom E (1982) Transepithelial invasion and intramesenchymal infiltration of the chick embryo chorioallantois by tumor cell lines. Cancer Res 42,1826–1837. Awwad JT, Sayegh RA, Tao XJ, Hassan T, Awwad ST and Isaacson K (1999) The SCID mouse: an experimental model for endometriosis. Hum Reprod 14,3107–3111. Barrier BF, Bates GW, Leland MM, Leach DA, Robinson RD and Propst AM (2004) Efficacy of anti-tumor necrosis factor therapy in the treatment of spontaneous endometriosis in baboons. Fertil Steril 81,775–779. Beliard A, Noel A, Goffin F, Frankenne F and Foidart JM (2002) Role of endo- crine status and cell type in adhesion of human endometrial cells to the peritoneum in nude mice. Fertil Steril 78,973–978. Bergqvist A, Jeppsson S, Kullander S and Ljungberg O (1985) Human uterine endometrium and endometriotic tissue transplanted into nude mice. Mor- phologic effects of various steroid hormones. Am J Pathol 121,337–341. Berkley KJ, Cason A, Jacobs H, Bradshaw H and Wood E (2001) Vaginal hyperalgesia in a rat model of endometriosis. Neurosci Lett 306,185–188. Berkley KJ, Dmitrieva N, Curtis KS and Papka RE (2004) Innervation of ectopic endometrium in a rat model of endometriosis. Proc Natl Acad Sci USA 101,11094–11098. Bruner KL, Matrisian LM, Rodgers WH, Gorstein F and Osteen KG (1997) Suppression of matrix metalloproteinases inhibits establishment of ectopic lesions by human endometrium in nude mice. J Clin Invest 99,2851–2857. Bruner KL, Eisenberg E, Gorstein F and Osteen KG (1999) Progesterone and transforming growth factor-beta coordinately regulate suppression of endometrial matrix metalloproteinases in a model of experimental endometriosis. Steroids 64,648–653. Bruner-Tran KL, Eisenberg E, Yeaman GR, Anderson TA, McBean J and Osteen KG (2002) Steroid and cytokine regulation of matrix metallopro- teinase expression in endometriosis and the establishment of experimental endometriosis in nude mice. J Clin Endocrinol Metab 87,4782–4791. Bulun SE, Zeitoun K, Takayama K, Noble L, Michael D, Simpson E, Johns A, Putman M and Sasano H (1999) Estrogen production in endometriosis and use of aromatase inhibitors to treat endometriosis. Endocr Relat Cancer 6,293–301. Cason AM, Samuelsen CL and Berkley KJ (2003) Estrous changes in vaginal nociception in a rat model of endometriosis. Horm Behav 44,123–131. Cummings AM and Metcalf JL (1995) Induction of endometriosis in mice: a new model sensitive to estrogen. Reprod Toxicol 9,233–238. Cummings AM and Metcalf JL (1996) Effect of surgically induced endometri- osis on pregnancy and effect of pregnancy and lactation on endometriosis in mice. Proc Soc Exp Biol Med 212,332–337. Cummings AM, Metcalf JL and Birnbaum L (1996) Promotion of endome- triosis by 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats and mice: time- dose dependence and species comparison. Toxicol Appl Pharmacol 138,131–139. D’Antonio M, Martelli F, Peano S, Papoian R and Borrelli F (2000) Ability of recombinant human TNF binding protein-1 (r-hTBP-1) to inhibit the development of experimentally-induced endometriosis in rats. J Reprod Immunol 48,81–98. DeVito MJ, Birnbaum LS, Farland WH and Gasiewicz TA (1995) Compari- sons of estimated human body burdens of dioxinlike chemicals and TCDD body burdens in experimentally exposed animals. Environ Health Per- spect 103,820–831. D’Hooghe TM (1997) Clinical relevance of the baboon as a model for the study of endometriosis. Fertil Steril 68,613–625. D’Hooghe TM and Debrock S (2002) Endometriosis, retrograde menstruation and peritoneal inflammation in women and in baboons. Hum Reprod Update 8,84–88. D’Hooghe TM, Bambra CS, Cornillie FJ, Isahakia M and Koninckx PR (1991) Prevalence and laparoscopic appearance of spontaneous endometriosis in the baboon (Papio anubis, Papio cynocephalus). Biol Reprod 45,411–416. D’Hooghe TM, Bambra CS, Suleman MA, Dunselman GA, Evers HL and Koninckx PR (1994) Development of a model of retrograde menstruation in baboons (Papio anubis). Fertil Steril 62,635–638. D’Hooghe TM, Bambra CS, Raeymaekers BM, De Jonge I, Hill JA and Koninckx PR (1995a) The effects of immunosuppression on development and progression of endometriosis in baboons (Papio anubis). Fertil Steril 64,172–178. D’Hooghe TM, Bambra CS, Raeymaekers BM, De Jonge I, Lauweryns JM and Koninckx PR (1995b) Intrapelvic injection of menstrual endometrium causes endometriosis in baboons (Papio cynocephalus and Papio anubis). Am J Obstet Gynecol 173,125–134. D’Hooghe TM, Bambra CS, De Jonge I, Lauweryns JM and Koninckx PR (1996a) The prevalence of spontaneous endometriosis in the baboon (Papio anubis, Papio cynocephalus) increases with the duration of captiv- ity. Acta Obstet Gynecol Scand 75,98–101. D’Hooghe TM, Bambra CS, Raeymaekers BM, Riday AM, Suleman MA and Koninckx PR (1996b) The cycle pregnancy rate is normal in baboons with stage I endometriosis but decreased in primates with stage II and stage III– IV disease. Fertil Steril 66,809–813. D’Hooghe TM, Bambra CS, Xiao L, Peixe K and Hill JA (2001) Effect of menstruation and intrapelvic injection of endometrium on inflammatory parameters of peritoneal fluid in the baboon (Papio anubis and Papio cynocephalus). Am J Obstet Gynecol 184,917–925. D’Hooghe TM, Nugent NP, Cuneo S, Chai DC, Deer F, Debrock S, Kyama CM, Mihalyi A and Mwenda JM (2006) Recombinant human TNFRSF1A (r-hTBP1) inhibits the development of endometriosis in baboons: a pro- spective, randomized, placebo- and drug-controlled study. Biol Reprod 74,131–136. Dick EJ Jr, Hubbard GB, Martin LJ and Leland MM (2003) Record review of baboons with histologically confirmed endometriosis in a large estab- lished colony. J Med Primatol 32,39–47. Dinulescu DM, Ince TA, Quade BJ, Shafer SA, Crowley D and Jacks T (2005) Role of K-ras and Pten in the development of mouse models of endometri- osis and endometrioid ovarian cancer. Nat Med 11,63–70. Dunselman GA, Willebrand D, Land JA, Bouckaert PX and Evers JL (1989) A rabbit model of endometriosis. Gynecol Obstet Invest 27,29–33. Efstathiou JA, Sampson DA, Levine Z, Rohan RM, Zurakowski D, Folkman J, D’Amato RJ and Rupnick MA (2005) Nonsteroidal antiinflammatory drugs differentially suppress endometriosis in a murine model. Fertil Steril 83,171–181. Eggermont J, Donnez J, Casanas-Roux F, Scholtes H and Van Langendonckt A (2005) Time course of pelvic endometriotic lesion revascularization in a nude mouse model. Fertil Steril 84,492–499. Elsheikh A, Milingos S, Loutradis D, Kallipolitis G and Michalas S (2003) Endometriosis and reproductive disorders. Ann N Y Acad Sci 997,247–254. Fang Z, Yang S, Lydon JP, DeMayo F, Tamura M, Gurates B and Bulun SE (2004) Intact progesterone receptors are essential to counteract the prolif- erative effect of estradiol in a genetically engineered mouse model of endometriosis. Fertil Steril 82,673–678. Fazleabas AT (2005) A baboon model for inducing endometriosis. Methods Mol Med 121,93–98. Fazleabas AT, Brudney A, Gurates B, Chai D and Bulun S (2002) A modified baboon model for endometriosis. Ann N Y Acad Sci 955,308–317; discus- sion 340–342, 396–406. Fazleabas AT, Brudney A, Chai D, Langoi D and Bulun SE (2003) Steroid receptor and aromatase expression in baboon endometriotic lesions. Fertil Steril 80(Suppl 2),820–827. Fortin M, Lepine M, Page M, Osteen K, Massie B, Hugo P and Steff AM (2003) An improved mouse model for endometriosis allows noninvasive assessment of lesion implantation and development. Fertil Steril 80(Suppl 2),832–838. Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022
  • 8. R.Grümmer 648 Fortin M, Lepine M, Merlen Y, Thibeault I, Rancourt C, Gosselin D, Hugo P and Steff AM (2004) Quantitative assessment of human endometriotic tis- sue maintenance and regression in a noninvasive mouse model of endometriosis. Mol Ther 9,540–547. Gashaw I, Hastings JM, Jackson KS, Winterhager E and Fazleabas AT (2006) Induced endometriosis in the baboon (Papio anubis) increases the expres- sion of the proangiogenic factor Cyr61 (CCN1) in eutopic and ectopic endometria. Biol Reprod 74,1060–1066. Gescher DM, Siggelkow W, Meyhoefer-Malik A and Malik E (2005) A priori implantation potential does not differ in eutopic endometrium of patients with and without endometriosis. Arch Gynecol Obstet 272,117–123. Giannopoulou E, Katsoris P, Hatziapostolou M, Kardamakis D, Kotsaki E, Polytarchou C, Parthymou A, Papaioannou S and Papadimitriou E (2001) X-rays modulate extracellular matrix in vivo. Int J Cancer 94,690–698. Giudice LC and Kao LC (2004) Endometriosis. Lancet 364,1789–1799. Golan A, Winston RM and Dargenio R (1984) Experimental endometriosis: a microsurgical animal model in rats. Isr J Med Sci 20,1094–1096. Greenberg LH and Slayden OD (2004) Human endometriotic xenografts in immunodeficient RAG-2/gamma(c)KO mice. Am J Obstet Gynecol 190,1788–1795; discussion 1795–1796. Grow DR, Williams RF, Hsiu JG and Hodgen GD (1996) Antiprogestin and/or gonadotropin-releasing hormone agonist for endometriosis treatment and bone maintenance: a 1-year primate study. J Clin Endocrinol Metab 81,1933–1939. Grummer R, Schwarzer F, Bainczyk K, Hess-Stumpp H, Regidor PA, Schindler AE and Winterhager E (2001) Peritoneal endometriosis: validation of an in-vivo model. Hum Reprod 16,1736–1743. Gruemmer R, Fechner S, Schwarzer F, Schmidt M and Winterhager E (2005) Expression of angiogenic factors and regulation of estrogen metabolising enzymes in human endometrial lesions grown in nude mice. Eur J Obstet Gynecol Reprod Med 123,S56 (abstract). Hadfield RM, Yudkin PL, Coe CL, Scheffler J, Uno H, Barlow DH, Kemnitz JW and Kennedy SH (1997) Risk factors for endometriosis in the rhesus monkey (Macaca mulatta): a case–control study. Hum Reprod Update 3,109–115. Halme J, Hammond MG, Hulka JF, Raj SG and Talbert LM (1984) Retrograde menstruation in healthy women and in patients with endometriosis. Obstet Gynecol 64,151–154. Hirata T, Osuga Y, Yoshino O, Hirota Y, Harada M, Takemura Y, Morimoto C, Koga K, Yano T, Tsutsumi O et al. (2005) Development of an experimen- tal model of endometriosis using mice that ubiquitously express green flu- orescent protein. Hum Reprod 20,2092–2096. Homm RJ, Garza DE, Mathur S, Austin M, Baggett B and Williamson HO (1989) Immunological aspects of surgically induced experimental endometriosis: variation in response to therapy. Fertil Steril 52,132–139. Hudelist G, Lass H, Keckstein J, Walter I, Wieser F, Wenzl R, Mueller R, Czerwenka K, Kubista E and Singer CF (2005) Interleukin 1alpha and tissue- lytic matrix metalloproteinase-1 are elevated in ectopic endometrium of patients with endometriosis. Hum Reprod 20,1695–1701. Hull ML, Charnock-Jones DS, Chan CL, Bruner-Tran KL, Osteen KG, Tom BD, Fan TP and Smith SK (2003) Antiangiogenic agents are effective inhibitors of endometriosis. J Clin Endocrinol Metab 88,2889–2899. Hull ML, Prentice A, Wang DY, Butt RP, Phillips SC, Smith SK and Charnock- Jones DS (2005) Nimesulide, a COX-2 inhibitor, does not reduce lesion size or number in a nude mouse model of endometriosis. Hum Reprod 20,350–358. Ingelmo JM, Quereda F and Acien P (1999) Intraperitoneal and subcutaneous treatment of experimental endometriosis with recombinant human interferon- alpha-2b in a murine model. Fertil Steril 71,907–911. Jones RC (1984) The effect of a luteinizing hormone releasing hormone (LRH) agonist (Wy-40,972), levonorgestrel, danazol and ovariectomy on experi- mental endometriosis in the rat. Acta Endocrinol (Copenh) 106,282–288. Katsuki Y, Takano Y, Futamura Y, Shibutani Y, Aoki D, Udagawa Y and Nozawa S (1998) Effects of dienogest, a synthetic steroid, on experimen- tal endometriosis in rats. Eur J Endocrinol 138,216–226. Kaufmann R, Rudolphi A, Boxberger HJ, Hainzl A, Rosenthal H and Reimann J (1995) Stable engraftment of human female genital mucous membrane xenografts on SCID mice. Gynecol Obstet Invest 40,97–100. Keenan JA, Williams-Boyce PK, Massey PJ, Chen TT, Caudle MR and Bukovsky A (1999) Regression of endometrial explants in a rat model of endometriosis treated with the immune modulators loxoribine and levami- sole. Fertil Steril 72,135–141. Kudoh M, Susaki Y, Ideyama Y, Nanya T, Mori M and Shikama H (1997) Inhibitory effects of a novel aromatase inhibitor, YM511, on growth of endometrial explants and insulin-like growth factor-I gene expression in rats with experimental endometriosis. J Steroid Biochem Mol Biol 63,75–80. Lapp T (2000) ACOG issues recommendations for the management of endometriosis. American College of Obstetricians and Gynecologists. Am Fam Physician 62,1431,1434. Lebovic DI, Kir M and Casey CL (2004) Peroxisome proliferator-activated receptor-gamma induces regression of endometrial explants in a rat model of endometriosis. Fertil Steril 82(Suppl 3),1008–1013. Maas JW, Groothuis PG, Dunselman GA, de Goeij AF, Struijker-Boudier HA and Evers JL (2001) Development of endometriosis-like lesions after transplantation of human endometrial fragments onto the chick embryo chorioallantoic membrane. Hum Reprod 16,627–631. MacKenzie WF and Casey HW (1975) Animal model of human disease. Endometriosis. Animal model: endometriosis in rhesus monkeys. Am J Pathol 80,341–344. Malik E, Meyhofer-Malik A, Berg C, Bohm W, Kunzi-Rapp K, Diedrich K and Ruck A (2000) Fluorescence diagnosis of endometriosis on the chorioallantoic membrane using 5-aminolaevulinic acid. Hum Reprod 15,584–588. Matsuura-Sawada R, Murakami T, Ozawa Y, Nabeshima H, Akahira J, Sato Y, Koyanagi Y, Ito M, Terada Y and Okamura K (2005) Reproduction of menstrual changes in transplanted human endometrial tissue in immuno- deficient mice. Hum Reprod 20,1477–1484. Matsuzaki S, Canis M, Darcha C, Dallel R, Okamura K and Mage G (2004) Cyclooxygenase-2 selective inhibitor prevents implantation of eutopic endometrium to ectopic sites in rats. Fertil Steril 82,1609–1615. Moon CE, Bertero MC, Curry TE, London SN, Muse KN, Sharpe KL and Vernon MW (1993) The presence of luteinized unruptured follicle syn- drome and altered folliculogenesis in rats with surgically induced endometriosis. Am J Obstet Gynecol 169,676–682. Nap AW, Groothuis PG, Demir AY, Maas JW, Dunselman GA, de Goeij AF and Evers JL (2003) Tissue integrity is essential for ectopic implantation of human endometrium in the chicken chorioallantoic membrane. Hum Reprod 18,30–34. Nap AW, Dunselman GA, de Goeij AF, Evers JL and Groothuis PG (2004) Inhibiting MMP activity prevents the development of endome- triosis in the chicken chorioallantoic membrane model. Hum Reprod 19,2180–2187. Nap AW, Dunselman GA, Griffioen AW, Mayo KH, Evers JL, Groothuis PG (2005) Angiostatic agents prevent the development of endometriosis-like lesions in the chicken chorioallantoic membrane. Fertil Steril 83,793–795. Nisolle M, Casanas-Roux F and Donnez J (2000) Early-stage endometriosis: adhesion and growth of human menstrual endometrium in nude mice. Fer- til Steril 74,306–312. Nothnick WB, Curry TE and Vernon MW (1994) Immunomodulation of rat endometriotic implant growth and protein production. Am J Reprod Immunol 31,151–162. Oosterlynck DJ, Meuleman C, Waer M, Koninckx PR and Vandeputte M (1993) Immunosuppressive activity of peritoneal fluid in women with endometriosis. Obstet Gynecol 82,206–212. Otsuka J, Okuda T, Sekizawa A, Amemiya S, Saito H, Okai T, Kushima M and Tachikawa T (2004) K-ras mutation may promote carcinogenesis of endometriosis leading to ovarian clear cell carcinoma. Med Electron Microsc 37,188–192. Pellicer A, Navarro J, Bosch E, Garrido N, Garcia-Velasco JA, Remohi J and Simon C (2001) Endometrial quality in infertile women with endometrio- sis. Ann N Y Acad Sci 943,122–130. Rajkumar K, Schott PW and Simpson CW (1990) The rat as an animal model for endometriosis to examine recurrence of ectopic endometrial tissue after regression. Fertil Steril 53,921–925. Rice VM (2002) Conventional medical therapies for endometriosis. Ann N Y Acad Sci 955,343–352; discussion 389–393, 396–406. Rier SE, Martin DC, Bowman RE, Dmowski WP and Becker JL (1993) Endometriosis in rhesus monkeys (Macaca mulatta) following chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Fundam Appl Toxicol 21,433–441. Rock JA, Prendergast RA, Bobbie D, Green WR, Parmley TH and Dubin NH (1993) Intraocular endometrium in the rabbit as a model for endometrio- sis. Fertil Steril 59,232–235. Rossi G, Somigliana E, Moschetta M, Santorsola R, Cozzolino S, Filardo P, Salmaso A and Zingrillo B (2000) Dynamic aspects of endometriosis in a mouse model through analysis of implantation and progression. Arch Gynecol Obstet 263,102–107. Saito T, Yoshizawa M, Yamauchi Y, Kinoshita S, Fujii T, Mieda M, Sone H, Yamamoto Y and Koizumi N (2003) Effects of the novel orally active Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022
  • 9. Animal models in endometriosis research 649 antiestrogen TZE-5323 on experimental endometriosis. Arzneimittelfor- schung 53,507–514. Sakata M, Terakawa N, Mizutani T, Tanizawa O, Matsumoto K, Terada N and Sudo K (1990) Effects of danazol, gonadotropin-releasing hormone agonist, and a combination of danazol and gonadotropin-releasing hormone agonist on experimental endometriosis. Am J Obstet Gynecol 163,1679–1684. Sampson JA (1927) Perotoneal endometriosis due to the menstrual dissemina- tion of endometrial tissue into the peritoneal cavity. Am J Obstet Gynecol 14,422–425. Sato N, Tsunoda H, Nishida M, Morishita Y, Takimoto Y, Kubo T and Noguchi M (2000) Loss of heterozygosity on 10q23.3 and mutation of the tumor suppressor gene PTEN in benign endometrial cyst of the ovary: possible sequence progression from benign endometrial cyst to endometrioid carcinoma and clear cell carcinoma of the ovary. Cancer Res 60,7052–7056. Schenken RS and Asch RH (1980) Surgical induction of endometriosis in the rabbit: effects on fertility and concentrations of peritoneal fluid prostag- landins. Fertil Steril 34,581–587. Schenken RS, Asch RH, Williams RF and Hodgen GD (1984) Etiology of infertility in monkeys with endometriosis: luteinized unruptured follicles, luteal phase defects, pelvic adhesions, and spontaneous abortions. Fertil Steril 41,122–130. Scher C, Haudenschild C and Klagsbrun M (1976) The chick chorioallantoic membrane as a model system for the study of tissue invasion by viral transformed cells. Cell 8,373–382. Schor E, Baracat EC, Simoes MJ, de Freitas V, Giannotti Filho O and de Lima GR (1999) Effects of conjugated estrogens and progestogen in surgically induced endometriosis in oophorectomized rats. Clin Exp Obstet Gynecol 26,158–161. Sharpe KL, Bertero MC, Muse KN and Vernon MW (1991) Spontaneous and steroid-induced recurrence of endometriosis after suppression by a gonadotropin-releasing hormone antagonist in the rat. Am J Obstet Gynecol 164,187–194. Somigliana E, Vigano P, Rossi G, Carinelli S, Vignali M and Panina- Bordignon P (1999) Endometrial ability to implant in ectopic sites can be prevented by interleukin-12 in a murine model of endometriosis. Hum Reprod 14,2944–2950. Steinleitner A, Lambert H and Roy S (1991a) Immunomodulation with pen- toxifylline abrogates macrophage-mediated infertility in an in vivo model: a paradigm for a novel approach to the treatment of endometriosis-associated subfertility. Fertil Steril 55,26–31. Steinleitner A, Lambert H, Suarez M, Serpa N, Robin B and Cantor B (1991b) Periovulatory calcium channel blockade enhances reproductive perform- ance in an animal model for endometriosis-associated subfertility. Am J Obstet Gynecol 164,949–952. Story L and Kennedy S (2004) Animal studies in endometriosis: a review. ILAR J 45,132–138. Tabibzadeh S, Miller S, Dodson WC and Satyaswaroop PG (1999) An experimental model for the endometriosis in athymic mice. Front Biosci 4,C4–C9. Te Linde RW and Scott RB (1950) Experimental endometriosis. Am J Obstet Gynecol 60,1147–1173. Uchiide I, Ihara T and Sugamata M (2002) Pathological evaluation of the rat endometriosis model. Fertil Steril 78,782–786. Van Langendonckt A, Casanas-Roux F, Eggermont J and Donnez J (2004) Characterization of iron deposition in endometriotic lesions induced in the nude mouse model. Hum Reprod 19,1265–1271. Vanden Heuvel JP, Clark GC, Tritscher A and Lucier GW (1994) Accumula- tion of polychlorinated dibenzo-p-dioxins and dibenzofurans in liver of control laboratory rats. Fundam Appl Toxicol 23,465–469. Vercellini P, Trecca D, Oldani S, Fracchiolla NS, Neri A and Crosignani PG (1994) Analysis of p53 and ras gene mutations in endometriosis. Gynecol Obstet Invest 38,70–71. Vernon MW and Wilson EA (1985) Studies on the surgical induction of endometriosis in the rat. Fertil Steril 44,684–694. Wheeler JM (1989) Epidemiology of endometriosis-associated infertility. J Reprod Med 34,41–46. Wolber EM, Kressin P, Meyhofer-Malik A, Diedrich K and Malik E (2003) Differential induction of matrix metalloproteinase 1 and 2 in ectopic endometrium. Reprod Biomed Online 6,238–243. Wright JA and Sharpe-Timms KL (1995) Gonadotropin-releasing hormone agonist therapy reduces postoperative adhesion formation and reformation after adhesiolysis in rat models for adhesion formation and endometriosis. Fertil Steril 63,1094–1100. Yang JZ, Agarwal SK and Foster WG (2000) Subchronic exposure to 2,3,7,8- tetrachlorodibenzo-p-dioxin modulates the pathophysiology of endometri- osis in the cynomolgus monkey. Toxicol Sci 56,374–381. Yano S, Ikegami Y and Nakao K (1996) Studies on the effect of the new non- steroidal aromatase inhibitor fadrozole hydrochloride in an endometriosis model in rats. Arzneimittelforschung 46,192–195. Yao Z, Shen X, Capodanno I, Donnelly M, Fenyk-Melody J, Hausamann J, Nunes C, Strauss J and Vakerich K (2005) Validation of rat endometriosis model by using raloxifene as a positive control for the evaluation of novel SERM compounds. J Invest Surg 18,177–183. Zaino RJ, Satyaswaroop PG and Mortel R (1985) Histologic response of nor- mal human endometrium to steroid hormones in athymic mice. Hum Pathol 16,867–872. Zamah NM, Dodson MG, Stephens LC, Buttram VC Jr, Besch PK and Kaufman RH (1984) Transplantation of normal and ectopic human endometrial tissue into athymic nude mice. Am J Obstet Gynecol 149,591–597. Zeitoun KM and Bulun SE (1999) Aromatase: a key molecule in the pathophys- iology of endometriosis and a therapeutic target. Fertil Steril 72,961–969. Zeitoun K, Takayama K, Sasano H, Suzuki T, Moghrabi N, Andersson S, Johns A, Meng L, Putman M, Carr B et al. (1998) Deficient 17beta- hydroxysteroid dehydrogenase type 2 expression in endometriosis: failure to metabolize 17beta-estradiol. J Clin Endocrinol Metab 83,4474–4480. Zhou HE and Nothnick WB (2005) The relevancy of the matrix metalloprotei- nase system to the pathophysiology of endometriosis. Front Biosci 10,569–575. Zondervan KT, Weeks DE, Colman R, Cardon LR, Hadfield R, Schleffler J, Trainor AG, Coe CL, Kemnitz JW and Kennedy SH (2004) Familial aggregation of endometriosis in a large pedigree of rhesus macaques. Hum Reprod 19,448–455. Submitted on March 15, 2006; resubmitted on May 5, 2006; accepted on May 20, 2006 Downloaded from https://academic.oup.com/humupd/article/12/5/641/779916 by guest on 28 January 2022