Download Endometrial dendritic cell populations during the normal menstrual

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Monoclonal antibody wikipedia , lookup

T cell wikipedia , lookup

DNA vaccination wikipedia , lookup

Hygiene hypothesis wikipedia , lookup

Phagocyte wikipedia , lookup

Immune system wikipedia , lookup

Lymphopoiesis wikipedia , lookup

Molecular mimicry wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Adaptive immune system wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Innate immune system wikipedia , lookup

Immunomics wikipedia , lookup

Transcript
doi:10.1093/humrep/den030
Human Reproduction Vol.23, No.7 pp. 1574– 1580, 2008
Advance Access publication on February 19, 2008
Endometrial dendritic cell populations during the normal
menstrual cycle
L. Schulke1, F. Manconi, R. Markham and I.S. Fraser
Department of Obstetrics and Gynaecology, Queen Elizabeth II Research Institute for Mothers and Infants, University of Sydney,
Sydney 2006, Australia
1
Correspondence address. E-mail: [email protected]
BACKGROUND: Dendritic cells (DCs) are specialized antigen presenting cells that are highly involved in the
stimulation and modulation of the immune response within mucosal surfaces, including the female reproductive
tract. DCs have been poorly characterized in the non-pregnant endometrium. METHODS: Hysterectomy specimens
were obtained from premenopausal women (n 5 49) with histoloigically normal endometrium. Endometrial sections
were stained immunohistochemically using antibodies for monoclonal mouse anti-human CD1a and CD83, two
markers which are specific for populations of immature and mature DCs, respectively. RESULTS: There was a significantly higher density of endometrial CD1a1 DCs than CD831 DCs throughout the menstrual cycle (P < 0.001).
The density of CD1a1 and CD831 DCs did not vary between the fundus and isthmus of the uterus. There was a significant increase in the density of CD1a1 DCs, but not CD831 DCs, in the basal layer of the endometrium through the
phases of the menstrual cycle. The density of CD831 was significantly greater in the basal layer compared with the
functional layer during both the proliferative (P 5 0.004) and secretory phases (P 5 0.001), whereas for CD1a1 DCs,
the greater density in the basal layer was only observed in the secretory phase (P < 0.001). CONCLUSIONS: The
highly coordinated cyclical changes in DC populations during the normal menstrual cycle reported in this study
may be important for local regulatory mechanisms relevant to menstruation and implantation; alterations in this
normal profile may contribute to the development of disturbances of function, fertility and even benign gynaecological
disease.
Keywords: dendritic cells; endometrium; menstrual cycle; CD1; CD83
Introduction
Dendritic cells (DCs) are a heterogeneous and dynamic
population of leukocytes that play an essential role in the
initiation and modulation of the immune response throughout
the human body, including the female reproductive tract.
DCs can be broadly classified according to their developmental
pathway; plasmacytoid (lymphoid) and myeloid DCs develop
from different haematopoietic progenitors and are proposed
to differ in terms of function, tissue distribution, cytokine production and markers. Plasmacytoid DCs are mainly involved in
the recognition of viruses and the production of type 1 interferon, whereas myeloid DCs, which are the focus of this
study, are known for their role in the initiation of the T-cell
response (Ueno et al., 2007).
In addition to the diversity in DC developmental pathways,
these cells are adaptive to pathogens and other environmental
signals and undergo phenotypic changes during their normal
lifespan. Myeloid DCs are integral to the maintenance of peripheral immunity and can be classified in terms of maturation
1574
status, which is associated with a specific functional profile
(Adams et al., 2005). Immature DCs patrol peripheral tissues
where they capture and process foreign antigens. In response
to danger signals such as foreign antigens or inflammatory
signals, DCs undergo maturation and travel to the lymph
nodes via the afferent lymphatics where antigens are presented
to T-cells in conjunction with major histocompatibility
complex (MHC) molecules. DCs can also regulate the development and maintenance of peripheral tolerance through the
uptake of self-antigens (Banchereau and Steinman, 1998).
Uterine leukocytes play a crucial role in the maintenance
of the normally sterile uterine environment during the menstrual cycle. Numerous studies have examined lymphocyte
populations within the normal endometrium during the menstrual cycle, but an examination of DCs has been overlooked
in this research (Kamat and Isaacson, 1987; Bulmer et al.,
1988; Starkey et al., 1991; Flynn et al., 2000). As in other
tissues, the uterine immune system is dependent on antigen
presenting cells (APCs) to initiate and stimulate a
# The Author 2008. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology.
All rights reserved. For Permissions, please email: [email protected]
Dendritic cells in the endometrium
co-ordinated immune response. DCs communicate both
directly and indirectly with many other types of leukocytes,
including T cells, B cells, natural killer cells and macrophages, thereby shaping the immune response in its entirety
(Banchereau and Steinman, 1998). Despite the importance
of DCs as antigen-presenting cells, there is a scarcity of
research examining DCs in the non-pregnant uterus during
the normal menstrual cycle.
The human uterus is unique in its ability to support the semiallograft fetus, while maintaining the immune protection that is
normally associated with other mucosal barriers within the
human body (Robertson, 2000). Previous studies have
focused on DCs in the decidualized endometrium and their
possible role in early spontaneous abortion (Kammerer et al.,
2000; Gardner and Moffett, 2003; Askelund et al., 2004).
DCs are crucial mediators of the immune response and tolerance, and are likely to be involved in this balance between
immune defence and fetal tolerance.
Menstruation has been described as an inflammatory event,
which is accompanied by an increase in the number of leukocytes, such as natural killer cells, macrophages and neutrophils, and their associated pro-inflammatory products
(Salamonsen and Lathbury, 2000). Although steroid hormones are largely responsible for the transformation of the
endometrium during the menstrual cycle, these immune
cells and their secreted products are potential mediators of
local endometrial changes (Jones et al., 2004). Chemokines
and cytokines, which are secreted by DCs and other leukocytes, have been implicated in the processes of menstruation,
tissue repair, angiogenesis, embryo implantation and parturition (Kayisli et al., 2002). The potential involvement of DCs
in the inflammatory environment of the uterus in the perimenstrual period is unknown. Alterations in the DC populations may disrupt the normal co-ordination of uterine leukocytes contributing to menstrual disturbances, or to the
development of pathology or infertility.
Our aim in this study was to characterize myeloid DCs
within the normal premenopausal endometrium during different phases of the menstrual cycle. We used the immunohistochemical markers CD1a and CD83, two markers which are
specific for immature and mature DCs, respectively, to quantify
DCs during the proliferative, secretory and menstrual phases of
the menstrual cycle. CD1a, a cell surface glycoprotein, that is
structurally related to the MHC molecules and which mediates
a MHC-independent antigen presentation pathway, is a highly
specific and sensitive marker of immature DCs and Langerhans
cells, a population of DCs found in the skin (Krenacs et al.,
1993). CD83 is a glycoprotein member of the immunoglobulin
superfamily that is strongly up-regulated during DC maturation
making it a robust marker for mature DCs (Lechmann et al.,
2002). In addition, we examined the density of DCs in different
regions of the uterus, the fundus and isthmus, and in different
layers of the endometrium, the functional and basal. An understanding of uterine DC populations may reveal important information about their role in maintenance of the normal immune
protection of the uterus during the menstrual cycle, as well as in
the processes of menstruation, implantation and in the development of disease or infertility.
Materials and Methods
Collection of tissue
This study was approved by the Human Ethics Committees of the
Southwest Sydney Area Health Service and the University of Sydney.
Hysterectomy samples were obtained from 49 premenopausal
women (mean age 45.0, range 36–51) with histologically normal
endometrium and pathology which was expected to have minimal
effect on endometrial immune function. Paraffin embedded fullthickness hysterectomy samples, containing both endometrium and
myometrium were obtained from recent surgical pathology archive
blocks. Indications for hysterectomy in these women included
uterine prolapse, heavy menstrual bleeding, small intramural fibroids
and minimal adenomyosis. None of the patients with uterine prolapse
had a complete prolapse (procidentia).
Menstrual cycle staging, using an idealized 28 day cycle, was determined by a blinded specialist gynaecological pathologist for all
samples. Uterine samples were collected from both the fundus and
isthmus of the uterus. Of the 28 normal samples collected from the
fundus, 6 were in the menstrual phase (days 1–7), 12 were in the proliferative phase (days 8–14), and 10 were in the secretory phase (days
15– 28). Of the 21 normal samples collected from the isthmus, 6 were
in the menstrual phase, 6 were in the proliferative phase, and 9 were in
the secretory phase.
Paired samples, collected from fundus and isthmus of the same
patient during the proliferative (six pairs), secretory (nine pairs) and
menstrual phases (six pairs) were used for the analysis of the effect
of region. Additional samples were obtained from the fundus only
and were combined with paired samples in subsequent analyses.
Immunohistochemistry
Tissue sections were cut at 4 mm and immunostained using monoclonal antibodies for mouse anti-human CD1a (dilution 1:100; Dako
Cytomation, Carpinteria, CA, USA) and mouse anti-human CD83
(dilution 1:50; Serotec, Raleigh, NC, USA) for 30 min at room temperature. Antigen retrieval was performed prior to immunostaining for
all sections using a pH 9 target retrieval solution (Dako Australia) for
all CD1a slides and Proteinase K (Dako Australia) was used for all
CD83 slides. Dual endogenous enzyme block (Dako Australia) was
applied to all sections for 10 min prior to incubation with the
primary antibody. Following the incubation with the primary antibodies, sections were washed in Tris Buffer and incubated with
REAL Link Biotinylated Secondary antibody (Dako Australia) for
15 min. Slides were then washed and incubated with streptavidin alkaline phosphatase (Dako Australia) for 15 min and stained with permanent fast red chromogen (Dako Australia) for 10 min. Slides were
counterstained with Mayers haemotoxylin prior to evaluation. Negative and positive control slides were incorporated into each slide
run. Skin and tonsil were used as positive control tissues for CD1a
and CD83, respectively. The negative control slides were processed
using the same protocol as sample slides, except the primary antibody
was omitted.
All immunostaining was carried out on a Dako Autostainer Model
S3400 (Dako USA). Images of the sections were captured using an
Olympus BX51 microscope and DP70 digital camera (Olympus,
Tokyo, Japan). DC staining was assessed using the Image Pro Plus Discovery software (MediaCybernetics, MD, USA). Images were obtained
from the functional and basal layers of the endometrium. Twenty
250 mm2 sections from both the functional and basal layers were captured for each tissue section. As the functional layer of the endometrium
is partially shed during menstruation, data were only obtained from the
basal layer for all samples for the menstrual phase. Cells were only
counted as positive if they demonstrated both positive staining and
1575
Schulke et al.
DC morphology. DC morphology was defined nucleated cells of appropriate size with visible processes extending from the cell body. A
visible DC process without an apparent cell body was not counted as
a DC. The density of CD1aþ and CD83þ DCs per mm2 of endometrium was calculated for each specimen.
Statistical analysis
Results were expressed as the mean + SE of CD1aþ or CD83þ DCs
per mm2. SPSS 13.0 (SPSS Inc., Chicago, IL, USA) was used to
perform all statistical analyses and to calculate means and standard
deviations.
The distribution of the data was examined prior to analysis and the
Student’s t-test was used to compare two normally distributed groups.
The Mann–Whitney U-test was used to compare two groups that were
not normally distributed. The Kruskal–Wallis test was used to
compare three skewed groups. The Paired t-test was used to
compare paired, normally distributed data. The Wilcoxon signed
ranks test was used for paired, skewed data. Statistical significance
was established at P-values of ,0.05.
Results
Hysterectomy samples from 49 premenopausal women were
obtained for this study. CD1aþ and CD83þ DCs were
present in the functional and basal layers of the endometrium
in all examined specimens (Fig. 1). There was a significantly
higher density of CD1aþ DCs than CD83þ DCs in both
layers of the endometrium throughout the menstrual cycle
(P , 0.001) (Table I).
Paired samples were collected from the fundus and isthmus
in order to examine the effect of region of the uterus on DC
density (Fig. 2). The density of CD1aþ DCs did not vary
between the fundus and the isthmus during the proliferative,
secretory and menstrual phases. There were also no significant
differences in the density of CD83þ DCs between the fundus
and isthmus of the uterus across all phases of the menstrual
cycle. Data from the paired fundal and the isthmic samples
were combined with unpaired samples for all further analyses.
The mean density of DCs in the functional and basal layers
was compared during the proliferative and secretory phases of
the menstrual cycle (Fig. 3). During the proliferative phase, the
concentration of CD1aþ DCs was similar in the functional and
basal layers of the endometrium. The total number of CD1aþ
DCs per mm2 was significantly higher in the basal layer than in
the functional layer during the secretory phase (P , 0.001).
The mean density of CD83þ DCs was significantly greater
in the basal layer than in the functional layer during both the
proliferative (P ¼ 0.004) and secretory phases (P ¼ 0.001).
The number of CD1aþ DCs significantly increased across
the phases of the menstrual cycle in the basal layer (Fig. 4).
The density of CD1aþ DCs increased in the basal
layer between the proliferative phase and the secretory phase
(P , 0.001), with a large influx of CD1aþ DCs occurring
during the menstrual phase (P ¼ 0.001). In the functional
Figure 1: CD1aþ and CD83þ DCs in the endometrium.
(A) CD1aþ DCs in the endometrium. (B) CD83þ DCs in the endometrium. All images are shown at 40 magnification and stained with the
chromogen permanent fast red
Table I. Mean density of CD1aþ and CD83þ DCs in the fundus and isthmus of the uterus and functional and basal endometrial layers during the menstrual
cycle.
Antibody
CD1a
CD83
Region
Fundus
Isthmus
Fundus
Isthmus
Proliferative
Menstrual
Functional
Basal
Functional
Basal
Basal
18.5 + 1.8
17.9 + 3.4
7.9 + 1.2
10.9 + 1.8
18.9 + 1.8
19.3 + 4.2
12.3 + 1.2
14.9 + 1.5
16.2 + 2.2
15.6 + 2.4
9.4 + 1.2
10.8 + 1.5
29.0 + 4.4
36.8 + 3.6
15.1 + 1.5
15.6 + 2.0
74.1 + 10.2
60.8 + 15.2
16.7 + 2.9
21.6 + 4.8
Data are represented by mean density (cells/mm2) + SEM.
1576
Secretory
Dendritic cells in the endometrium
Figure 2: Comparison of the mean density of CD1aþ (A) and CD83þ (B) endometrial DCs in paired samples taken from the fundus and isthmus
of the uterus during the menstrual cycle.
Data are represented by mean + SEM. Graphs A and B use different scales
Figure 3: Comparison of the mean density of CD1aþ (A) and CD83þ (B) DCs in the functional and basal layers of the endometrium during the
menstrual cycle.
Data are represented by mean + SEM. Graphs A and B use different scales. Significant difference versus functional: *P , 0.001; **P ¼ 0.004;
***P ¼ 0.001
layer, the number of CD1aþ DCs remained stable between the
proliferative and secretory phases. There was a small and progressive increase in CD83þ DCs during the menstrual cycle,
but this increase was not statistically significant.
Discussion
This study utilized immunohistochemical methods to demonstrate that uterine DC populations vary by phase of the
menstrual cycle and layer of the endometrium, but not by
region of the uterus. Overall, the endometrial density of both
CD1aþ and CD83þ DCs was relatively low compared with
other types of leukocytes, such as macrophages and natural
killer cells (Bulmer et al., 1988). Only a small number of
DCs are necessary to patrol peripheral tissues and trigger
a strong T-cell response in the presence of foreign antigen
(Banchereau and Steinman, 1998). Previous research found
few to no DCs in the endometrium using early DC markers
1577
Schulke et al.
Figure 4: Comparison of the mean density of uterine CD1aþ (A) and CD83þ (B) DCs during the menstrual cycle.
Data are represented by mean + SEM. Graphs A and B use different scales. Significant difference compared with the preceding phase: *P , 0.001
and **P ¼ 0.001
(Kamat and Isaacson, 1987; Bulmer et al., 1988). The relative
rarity of DCs and the lack of robust immunohistochemical
markers made it previously difficult to quantify uterine DCs.
We also found a significantly lower density of mature uterine
DCs than immature uterine DCs which supports previous findings (Rieger et al., 2004). DC maturation is linked with
migration out of peripheral tissues to secondary lymphoid
organs, so the number of mature DCs in the uterus at any one
time is likely to be small (Banchereau et al., 2000). The
density of CD83þ DCs was higher in the basal layer during
both the proliferative and secretory phases, which is also consistent with the concept that these mature DCs are transiting out
of the uterus.
CD1aþ immature DCs increased in density throughout the
menstrual cycle in the basal layer of the endometrium,
whereas the number of CD83þ mature DCs remained relatively constant throughout the menstrual cycle. The increase
in immature CD1aþ DCs, but not mature CD83þ DCs
throughout the menstrual cycle suggests that the influx of
DCs into the uterus may be hormonally regulated, but that
DC maturation is not influenced by hormones. King et al.
(1996) evaluated the expression of estrogen and progesterone
receptor in CD45þ endometrial lymphocytes and found that
neither receptor was expressed by these cells. It is likely that
the influence of steroid hormones on uterine leukocytes including DCs is indirect via actions on cytokines and chemokines, a
family of cytokines whose major known function is chemotaxis
of leukocytes (McColl, 2002).
We found that the density of immature CD1aþ DCs in the
basal layer of the endometrium increased throughout the menstrual cycle, presumably as DCs migrated into the uterus. The
withdrawal of progesterone at the end of the luteal phase
1578
coincides with an uterine inflammatory response, which is
accompanied by an increase in other types of leukocytes,
such as macrophages, neutrophils and natural killer cells (Salamonsen and Lathbury, 2000). This study indicated that immature DCs, like other uterine leukocytes, respond to the
hormonal changes within the uterus and migrate into the
uterus during the secretory and menstrual phases.
Using a different marker for immature DCs, Rieger et al.
(2004) found that the density of immature DCs positive for
DC-SIGN (DC-specific ICAM-grabbing nonintegrin, classified
as CD209) did not change during the proliferative and
secretory phases of the menstrual cycle. Hysterectomy
samples from the menstrual phase were not included in this
analysis and the density of DC-SIGNþ DCs may not have
increased until the menstrual phase. It is also possible that
the influx of certain immature DC populations into the uterus
is hormone dependent, whereas the migration of other types
of DCs is regulated by other mechanisms. Further studies utilizing additional DC markers are required to fully characterize
this heterogeneous population of uterine DCs.
The exact role of DCs during the peri-menstrual period is
unknown, but DCs can produce matrix metalloproteinases
(MMPs), cytokines and chemokines, which are likely to influence the local uterine environment in the period leading up to
menstruation or implantation. DCs in the skin were found to
produce MMP-9 and MMP-2, which likely play a functional
role in DC migration (Osman et al., 2002; Ratzinger et al.,
2002). MMPs produced by uterine DCs could be involved in
tissue breakdown at menstruation.
DCs can also produce cytokines and chemokines in response
to environmental triggers, such as inflammatory stimuli
(Bengtsson et al., 2004). Gene expression profiling studies
Dendritic cells in the endometrium
have identified a number of cytokines and chemokines produced by DCs during different stages of maturation, including
the interleukins, IL-10, IL-12, IL-6, TNF-a (tumour necrosis
factor-a), RANTES (regulated upon activation normal T-cell
expressed and secreted protein) and MCP-1 (monocyte chemotactic protein-1) (Nagorsen et al., 2004). The production of
cytokines and chemokines by DCs not only regulates the
uterine local environment, but also other leukocyte populations
and their secreted products. The careful co-ordination of these
cytokines and chemokines is important in the regulation of
uterine function and disturbances in the normal profile may
contribute to implantation defects or infertility (Kayisli et al.,
2002).
The mucosal lining of the female reproductive tract depends
on the innate immune system to maintain a barrier against
encountered foreign antigens, such as micro-organisms and
semen. DCs can directly recognize specific pathogens using
toll-like receptors, a class of pattern-recognition receptors
and initiate an immune response, providing an important link
between adaptive and innate immunity (Pasare et al., 2004).
The need for immune protection against microbial infection
and the acquisition of sexually transmitted disease is more substantial in the lower genital tract than in the uterus, and correspondingly, there is a high mean density of immature DCs in
the lower genital tract, specifically in the vagina (Pudney
et al., 2005), ectocervix (Morelli et al., 1992) and uterine
cervix (Poppe et al., 1998). Therefore, it would be expected
that the density of uterine DCs would be greater in the
isthmic region than in the fundus. Surprisingly, we found no
significant differences between the density of CD1aþ and
CD83þ DCs in the fundus and isthmus of the uterus. DCs
are highly motile and migrate in response to local signals; it
is likely that under normal conditions uterine DCs are evenly
distributed throughout the uterus and density can be altered
in the presence of pathogens or other signals.
This study utilized tissue samples from women undergoing
hysterectomy for a number of indications, but who had histologically normal endometrium. All women who have a hysterectomy are likely to have some uterine abnormality that
precludes them from being completely ‘normal’. Also,
women undergoing hysterectomy are likely to be in their late
30s or 40s, at the end of their reproductive lifetime. An
attempt was made to exclude any patients with any conditions
or pathologies that affect the endometrium or immune system.
Although this study was not designed to examine DC populations in a number of minor pathologies, statistical analysis
revealed no significant or suggestive differences between the
subgroups.
In conclusion, we demonstrated that the density of immature
CD1aþ DCs in endometrium increased throughout the menstrual cycle and is likely to be indirectly regulated by steroid
hormones. Variation in the density of mature CD83þ DCs
does not appear to be hormone dependent, and the overall
number of these mature DCs is small, presumably due to
their continual transit out of the endometrium upon maturation.
The endometrium is a highly dynamic tissue where changes
occur not only across the phases of the menstrual cycle, but
on a daily or even hourly basis especially around the time of
menstruation. DCs may be involved in the signalling and regulation of some of these local changes in the uterus. CD1aþ and
CD83þ DCs represent only a portion of the entire uterine DC
population and further studies are necessary to identify and
characterize all uterine DCs. The alteration of uterine DC
populations may contribute to the menstrual disturbances,
implantation defects, infertility or other pathologies which
characterize endometrial disease and further investigation is
needed to examine the possible role of uterine DCs in these
conditions.
Acknowledgements
The authors thank Professor Peter Russell (Department of Pathology,
University of Sydney, Australia) for his assistance in obtaining the
pathology blocks used in this study and for blindly reviewing the
tissue sections; Natsuko Tokushige (University of Sydney, Australia)
and Lawrence Young (Dako, Australia) for their technical support;
and Georgina Luscombe (University of Sydney, Australia) for her
statistical expertise.
Funding
This study was supported by research funding from the Department of Obstetrics and Gynaecology, University of Sydney.
References
Adams S, O’Neill DW, Bhardwaj N. Recent advances in dendritic cell biology.
J Clin Immunol 2005;25:87 –98.
Askelund K, Liddell HS, Zanderigo AM, Fernando NS, Khong TY, Stone PR,
Chamley LW. CD83(þ)dendritic cells in the decidua of women with
recurrent miscarriage and normal pregnancy. Placenta 2004;25:140– 145.
Banchereau J, Steinman RM. Dendritic cells and the control of immunity.
Nature 1998;392:245– 252.
Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B,
Palucka K. Immunobiology of dendritic cells. Annu Rev Immunol
2000;18:767– 811.
Bengtsson AK, Ryan EJ, Giordano D, Magaletti DM, Clark EA.
17beta-estradiol (E2) modulates cytokine and chemokine expression in
human. Blood 2004;104:1404–1410.
Bulmer JN, Lunny DP, Hagin SV. Immunohistochemical characterization of
stromal leucocytes in nonpregnant human endometrium. Am J Reprod
Immunol 1988;17:83–90.
Flynn L, Byrne B, Carton J, Kelehan P, O’Herlihy C, O’Farrelly C. Menstrual
cycle dependent fluctuations in NK and T-lymphocyte subsets from
non-pregnant human endometrium. Am J Reprod Immunol
2000;43:209– 217.
Gardner L, Moffett A. Dendritic cells in the human decidua. Biol Reprod
2003;69:1438–1446.
Jones RL, Hannan NJ, Kaitu’u Tu J, Zhang J, Salamonsen LA. Identification of
chemokines important for leukocyte recruitment to the human endometrium
at the times of embryo implantation and menstruation. J Clin Endocrinol
Metab 2004;89:6155– 6167.
Kamat BR, Isaacson PG. The immunocytochemical distribution of leukocytic
subpopulations in human endometrium. Am J Pathol 1987;127:66 –73.
Kammerer U, Schoppet M, McLellan AD, Kapp M, Huppertz HI, Kampgen E,
Dietl J. Human decidua contains potent immunostimulatory CD83(þ)
dendritic cells. Am J Pathol 2000;157:159– 169.
Kayisli UA, Mahutte NG, Arici A. Uterine chemokines in reproductive
physiology and pathology. Am J Reprod Immunol 2002;47:213– 221.
King A, Gardner L, Loke YW. Evaluation of oestrogen and progesterone
receptor expression in uterine mucosal lymphocytes. Hum Reprod
1996;11:1079–1082.
Krenacs L, Tiszalvicz L, Boumsell L. Immunohistochemical detection of
CD1A antigen in formalin-fixed and paraffin-embedded tissue sections
with monoclonal antibody 010. J Pathol 1993;171:99–104.
1579
Schulke et al.
Lechmann M, Berchtold S, Hauber J, Steinkasserer A. CD83 on dendritic cells:
more than just a marker for maturation. Trends Immunol 2002;23:273–275.
McColl SR. Chemokines and dendritic cells: a crucial alliance. Immunol Cell
Biol 2002;80:489–496.
Morelli AE, di Paola G, Fainboim L. Density and distribution of Langerhans
cells in the human uterine cervix. Arch Gynecol Obstet 1992;252:65 –71.
Nagorsen D, Marincola FM, Panelli MC. Cytokine and chemokine expression
profiles of maturing dendritic cells using multiprotein platform arrays.
Cytokine 2004;25:31–35.
Osman M, Tortorella M, Londei M, Quaratino S. Expression of matrix
metalloproteinases and tissue inhibitors of metalloproteinases define the
migratory characteristics of human monocyte-derived dendritic cells.
Immunology 2002;105:73– 82.
Pasare C, Medzhitov R, Pasare C, Medzhitov R. Toll-like receptors: linking
innate and adaptive immunity. Microbes Infect 2004;6:1382–1387.
Poppe WA, Drijkoningen M, Ide PS, Lauweryns JM, Van Assche FA.
Lymphocytes and dendritic cells in the normal uterine cervix. An
immunohistochemical study. Eur J Obstet Gynecol Reprod Biol
1998;81:277– 282.
Pudney J, Quayle AJ, Anderson DJ. Immunological microenvironments in the
human vagina and cervix: mediators of cellular immunity are concentrated in
the cervical transformation zone. Biol Reprod 2005;73:1253– 1263.
1580
Ratzinger G, Stoitzner P, Ebner S, Lutz MB, Layton GT, Rainer C, Senior RM,
Shipley JM, Fritsch P, Schuler G et al. Matrix metalloproteinases 9
and 2 are necessary for the migration of Langerhans cells and dermal
dendritic cells from human and murine skin. J Immunol
2002;168:4361–4371.
Rieger L, Honig A, Sutterlin M, Kapp M, Dietl J, Ruck P, Kammerer U.
Antigen-presenting cells in human endometrium during the
menstrual cycle compared to early pregnancy. J Soc Gynecol Invest
2004;11:488–493.
Robertson SA. Control of the immunological environment of the uterus. Rev
Reprod 2000;5:164–174.
Salamonsen LA, Lathbury LJ. Endometrial leukocytes and menstruation. Hum
Reprod Update 2000;6:16– 27.
Starkey PM, Clover LM, Rees MCP. Variation during the menstrual cycle of
immune cell populations in human endometrium. Eur J Obstet Gynecol
Reprod Biol 1991;39:203– 207.
Ueno H, Klechevsky E, Morita R, Aspord C, Cao T, Matsui T, Di Pucchio T,
Connolly J, Fay JW, Pascual V et al. Dendritic cell subsets in health and
disease. Immunol Rev 2007;219:118–142.
Submitted on June 27, 2007; resubmitted on January 14, 2008; accepted on
January 22, 2008