Open Access

Inflammatory cytokine profile of co‑cultivated primary cells from the endometrium of women with and without endometriosis

  • Authors:
    • Adriana Luckow Invitti
    • Eduardo Schor
    • Rafael Martins Parreira
    • Alexander Kopelman
    • Gil Kamergorodsky
    • Giovana Aparecida Gonçalves
    • Manoel João Batista Castello Girão
  • View Affiliations

  • Published online on: June 6, 2018     https://doi.org/10.3892/mmr.2018.9137
  • Pages: 1287-1296
  • Copyright: © Luckow Invitti et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endometriosis is a chronic gynecological disorder defined as the presence of endometrial tissue within extra-uterine sites. The primary symptoms are infertility and chronic pain. The inflammatory environment and aberrant immune responses in women with endometriosis may be directly associated with the initiation and progression of endometriotic lesions. In the present study, the secretion of inflammatory cytokines was evaluated in cultures of primary endometrial cells (ECs) isolated from the endometrium of women with and without endometriosis. The presence of endometriotic cells leads to alterations in the secretory profile of healthy ECs. The expression of the inflammatory cytokines interleukin (IL)‑6 and IL‑8 was significantly increased in endometriotic and co‑cultured cells compared with healthy ECs. IL‑6 expression was strongly correlated with IL‑8 expression in endometriotic cells. IL‑1β expression was increased on day 10 of co‑culture to 48.30 pg/ml and may be associated with the long‑term co‑culture, rather than IL‑6 and IL‑8 expression. IL‑6 expression was strongly correlated with cell number, whereas IL‑8 expression was moderately correlated with cell number. Additionally, it was observed that co‑cultured cells exhibited a different population of cells, with expression of the mesenchymal stem cell marker cell surface glycoprotein MUC18, indicating a putative role of endometrial mesenchymal stem cells in the secretion of cytokines and disease development. These results indicate a predominant role of primary endometriotic cells in the secretion of cytokines, which contributes to the disrupted peritoneal and endometrial environment observed in the women with endometriosis.

Introduction

Endometriosis is a chronic gynecological disorder defined as the presence of endometrial tissue within extra-uterine sites (1,2). It affects 10–15% of women of reproductive age and results in a markedly reduced quality of life (35). The primary symptoms are infertility and chronic pain (6,7). It is a hormone-dependent and chronic inflammatory disease (1,2,8), indicating that the endometrium and the peritoneal environment are directly associated with its pathogenesis.

The progression of endometriosis is dependent on genetic, endocrine, immunological and environmental factors (9,10). The endometrium, serum and peritoneal fluid of women with endometriosis have abnormal levels of inflammatory cytokines, angiogenic, growth and adhesion factors, and cancer-like molecules (6,11,12). These soluble factors are thought to have predominant involvement in disease initiation and progression. Interleukin (IL)-1β has a proliferative effect on endometriotic cells that does not occur in healthy endometrial cells (13). IL-1β stimulates the production of IL-6 and IL-8 in endometriotic cell cultures, which further induce proliferation (14,15) and reduce the apoptotic rate (1619). Additionally, IL-1β increases the shedding of intercellular adhesion molecule (ICAM)-1 from peritoneal mesothelial cells, indicating a role in the neovascularization mediated by IL-6 and vascular endothelial growth factor (VEGF) (20). Furthermore, higher levels of IL-1β may indicate the conversion of inflammation from an acute to a chronic form (21).

Tumor necrosis factor (TNF)-α, IL-6, IL-8, IL-10, VEGF and C-C motif chemokine 2 (MCP-1) expression is increased in the peritoneal fluid of women with endometriosis (11,18,2124). TNF-α is associated with pluripotency mediation and inflammatory cytokine production, particularly IL-8, in endometriotic tissues (25). IL-8 increases the adhesion of endometrial stroma to extracellular matrix proteins, in addition to increasing metalloproteinase expression and proliferation (2628). IL-8 may be a key cytokine in the progression of endometriotic lesions, by stimulating growth and indirectly protecting implants against apoptosis (18). IL-6 is secreted by endometrial cells and may have an important role in the pathology of endometriosis with interferon-γ. IL-6 increases macrophage expression of ICAM-1 in patients with endometriosis (29,30). Increased peritoneal concentrations of IL-6 and IL-8 are associated with different stages of the disease (23,31). In addition, the resistance to progesterone observed in endometriosis (3234) is linked to the aberrant expression of cytokines (35,36).

The aberrant peritoneal environment, in addition to discrepancies in apoptosis and proliferation, result in abnormal immune cell clearance in women with endometriosis, providing a longer survival of the endometrial cells regurgitated by uterine tubes and the establishment of endometrial implants (5,24,31,37). Recently, the evaluation of peritoneal cytokines in patients with endometriosis indicated that the inflammatory environment observed in these patients is triggered by the establishment and growth of endometrial implants (38). Therefore, the present study investigated inflammatory cytokine secretion in the culture media of healthy and endometriotic primary cells. It was hypothesized that co-culture with endometriotic cells may lead to inflammatory cytokine secretion and phenotypic modifications in healthy endometrial cells associated with endometriosis implant establishment.

Materials and methods

Human tissues and cell culture

The protocol of the present study was approved by the Federal University of São Paulo ethical committee (registration no. 1044/11, São Paulo, Brazil). Written informed consent was obtained from all patients. Endometrial cells (ECs) from healthy individuals and patients with endometriosis were obtained from the Endometrium and Endometriosis Cell Bank of the Pelvic Pain and Endometriosis Unit of the Federal University of São Paulo. The selected samples were collected from August 2013 to April 2014. Human endometrium samples were collected from fertile cycling women aged 18–45 undergoing laparoscopic surgery for endometriosis stage IV (endometriosis group; n=3) and tubal ligation (control group; n=3). Patients had not taken exogenous hormones, given birth or breastfed for 3 months prior to surgery; patients with co-morbidities including teratoma, endometrial polyps or any other proliferative disease were excluded. The collected endometrial tissue was separated into two for either histological analysis (39,40) (data not shown) or cell culture.

The tissue for cell culture was immediately placed in Dulbecco's modified Eagle's medium with nutrient mixture F12 (DMEM/F12; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 400 U/ml penicillin and 400 µg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc.), stored at 4°C and processed within 2–24 h. The endometrial tissue was dissociated with 255 units of collagenase type IA (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and 3 units of dispase (Gibco; Thermo Fisher Scientific, Inc.) in DMEM/F12 and was subsequently incubated for 40 min in a 37°C water bath under constant agitation. The cell suspension obtained was centrifuged at 500 × g for 5 min at room temperature and the pellet was resuspended in 5 ml EC medium containing DMEM/F12 at pH 7.4, 1% Minimum Essential Medium non-essential amino acids (Gibco; Thermo Fisher Scientific, Inc.), 0.1 mmol/l 2-mercaptoethanol (Sigma-Aldrich; Merck KGaA), 100 µ/ml penicillin and 100 µ/ml streptomycin and 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), and inoculated in a pre-coated 25 cm2 cell culture flask. The cells were grown in EC medium supplemented with 10% FBS at 37°C until approximately 70% confluence was reached. At this stage, cells were subcultured. The cells were stored in liquid nitrogen.

Healthy and endometriotic cell co-culture

Co-cultures were performed in 12-well culture plates. Healthy ECs were pre-stained with 2 µg/ml Calcein-AM (Thermo Fisher Scientific, Inc., Waltham, MA, USA) for 30 min at 37°C with 5% CO2. Cell number and viability were assessed with a Countess™ Automated Cell Counter (Invitrogen; Thermo Fisher Scientific Inc.); 10 µl cell suspension was thoroughly mixed with 10 µl 0.4% Trypan Blue (Invitrogen; Thermo Fisher Scientific Inc.), then, 10 µl was added to the chamber slide, the cells were settled for 30 sec at room temperature prior to counting; according to the manufacturer's protocols. Healthy and endometriotic cells were mixed at a 1:1 ratio and seeded onto 12-well plates at a density of 5,000 cells/cm2 and cultured in EC medium supplemented with 10% FBS for 10 days. The adhesion of green (healthy) and unstained (endometriotic) cells was reported by light microscopy (magnification, ×50, Axio Observer, Zeiss GmbH, Jena, Germany) 24 h after the initial cell seeding, the stained and non-stained cells were visually counted in four different fields for each triplicate. The culture medium was changed every 2 days and in the day prior to each control point (1st, 3rd, 7th and 10th days of culture). Cells were harvested during the 1st, 3rd, 7th and 10th days of culture, suspended in FBS with 10% dimethyl sulfoxide and stored in liquid nitrogen. The culture medium was collected and stored at −80°C on days 1, 3, 7 and 10 of culture. As a control, healthy and endometriotic cell groups were cultured separately as described above.

Culture medium cytokines detection

IL-1β, IL-6, IL-8, IL-10, IL-12p70 and TNF-α expression levels were detected in the supernatant culture medium samples of single cell or co-cultured cells at days 1, 3, 7 and 10 by flow cytometry using the phycoerythrin (PE)-conjugated beads from the Human Inflammatory Cytokine cytometric bead array kit (BD Biosciences, Franklin Lakes, NJ, USA). Events acquisition was performed in a CANTO II 6-colour flow cytometer (BD Biosciences) and data were analyzed with FlowJo software (version 10.0.7; FlowJo LLC, Ashland, OR, USA).

Flow cytometry of membrane markers

Healthy and endometriotic cells prior to the co-culture seeding and co-cultured cells harvested in the 1st, 3rd, 7th and 10th days of culture were thawed on ice, centrifuged at 500 × g for 5 min 4°C, ressuspended in pH 7.4 1X PBS (Gibco; Thermo Fisher Scientific, Inc.) with 1% FBS (Gibco; Thermo Fisher Scientific, Inc.) for blocking and incubated with directly conjugated antibodies for the surface markers aminopeptidase N [CD13-allophycocyanin (APC); cat no. 555394; BD Biosciences] at 1:5 dilution ratio and cell surface glycoprotein MUC18 [CD146-PE; cat no. 561013; BD Biosciences] at 1:20 dilution ratio for 1 h at 4°C. Cells were washed with PBS 1X pH 7.4 and centrifuged at 500 × g for 5 min 4°C, ressuspended in PBS 1X pH 7,4 and subsequently analyzed with a BC FACSCanto II flow cytometer (BD Biosciences). The experiment controls were performed using imunoglubulin G1 isotype controls conjugated with PE (cat no. 550617) and APC (cat no. 550854) both at 1:5 working dilution ratio (BD Biosciences). Data were analyzed in FlowJo software (version 10.0.7; FlowJo LLC).

Statistical analysis

The data were analyzed with PASW Statistics 18.0.0 (IBM Corp., Armonk, NY, USA). All cell cultures were performed in experimental triplicates. Data are presented as the mean ± standard error (SE). Cytokine data were analyzed with repeated measures analysis followed by Dunnett's T3 pairwise comparisons post-hoc test between the groups that presented unequal variances. Cell marker data were analyzed with repeated measures analysis followed by a Bonferroni corrected pairwise comparisons test within-subjects factors. The curves obtained were tested for sphericity using Mauchly's test and within-subject effects were corrected using the Greenhouse-Geisser test when sphericity assumption was violated. Simple and multiple linear regression were performed to identify the correlation between the variables obtained, where 0< r <0.25 was null; 0.25 < r <0.50 was weakly correlated; 0.50< r <0.75 was moderately correlated; and 0.75 < r <1 was strongly correlated (41). P<0.05 was considered to indicate a statistically significant difference.

Results

Competitive relationship between healthy and endometriotic cells in co-culture systems

The adhesion of endometriotic (unstained) and healthy (Calcein-AM stained) cells in the first day of the co-culture is presented in Fig. 1. The amount of stained and not stained cells was visually semiquantitative evaluated in four microscopy fields in each triplicate. No observable difference was determined in the amount of endometriotic or healthy cells attached, indicating an equal number of each type of cell at the beginning of co-cultivation.

Inflammatory cytokine profile of healthy, endometriotic and co-cultured cells

The levels of inflammatory cytokines (IL-1β, IL-6, IL-8, IL-10, IL-12p70 and TNF-α) secreted into the culture medium were determined over time in endometriotic and healthy cells cultured alone, in addition to co-cultured cells. Insignificant levels of IL-10, IL12p70 and TNF-α were detected in the culture medium of all tested combinations of cells (data not shown). However, the IL-6 concentration increased over time in all groups (Fig. 2A). The variation observed for the endometriotic cells was similar to that observed in the co-cultured ECs (P=1.000, across all time points). The healthy ECs had a significantly different profile of IL-6 secretion, compared with co-cultured ECs (P=0.017, across all time points). As presented in Fig. 2, IL-6 secretion was higher in the endometriosis and co-culture groups. A strong correlation was detected between the number of cells and the concentration of IL-6 in the medium of healthy and endometriotic cells (Table I).

Table I.

Multiple correlation coefficient between inflammatory cytokine expression and cell number in endometriotic and healthy endometrial cells individually and co-cultured over time.

Table I.

Multiple correlation coefficient between inflammatory cytokine expression and cell number in endometriotic and healthy endometrial cells individually and co-cultured over time.

EndometriosisHealthyCo-cultured cells



PairsrP-valuerP-valuerP-value
No. of cells and IL-60.8711400.925280
No. of cells and IL-80.602550.00010.534070.0087
IL-6 and IL-8 (all time points)0.8421100.506480.01370.69470
IL-1β and IL-6 (day 10) 0.55650.1197
IL-1β and IL-8 (day 10) 0.21580.5771

[i] IL, interleukin.

Pro-angiogenic IL-8 was differentially secreted in healthy, endometriotic and co-cultured ECs (Fig. 2B). The most significant variation occurred from day 7, when the concentration increased in all groups. The differences between the curves of IL-8 concentration were statistically significant (P=0.010). The secreted IL-8 level in the healthy EC culture was significantly lower compared with the endometriosis (P=0.026, across all time points) and co-cultured (P=0.006, across all time points) groups. Similar to the trend observed for IL-6, the secretion of IL-8 was similar in the endometriotic and co-cultured cells (P=0.474, across all time points). The number of cells and IL-8 concentration were moderately correlated in healthy and endometriotic cells (Table I). Additionally, there was a moderate correlation between IL-6 and IL-8 secretion in the healthy and co-cultured cells. A strong correlation between the secretion of these cytokines was observed in the endometriotic cells (Table I).

The co-cultured cells had 48.30 pg/ml (SE, 9.68) IL-1β at day 10 of culture (data not shown); IL-1β levels at days 1, 3 and 7 were undetectable. The secretion of IL-1β appears to be associated with the effects of long-term co-culturing of the cells, although not directly to the secretion of IL-6 or IL-8 (Table I).

Population profile of co-cultured ECs

Flow cytometry analysis of the cells cultured alone and co-cultured cells using the size (forward scatter) and complexity (side scatter) parameters demonstrated the presence of two main populations in the samples: One and two. Population one represented <5% of the total events collected in the healthy (Fig. 3A) and endometriosis (Fig. 3B) samples alone (2.95%; SE, 0.44). The percentage of this population increased by 15-fold in the first day of co-culture (Day 1, 45.93%; SE, 3.58; P=0.000179 vs. cells cultured alone; Fig. 3C). No population differences were observed in the ECs individually cultured over time (data not shown).

Population one diminished at day 3 (32.35%; SE, 2.16; P=0.028412 vs. day 1 of co-culture; Fig. 3D) and day 7 (11.89%; SE, 2.33; P=0.000076 vs. day 3 of co-culture; Fig. 3E) of co-culture and appeared to stabilize between days 7 and 10 (Fig. 3F) of culture (10.40%; SE, 1.79; P=0.302732, day 7 vs. day 10 of co-culture).

Population two was more prevalent in the EC samples cultured alone (87.36%; SE, 1.52; Fig. 3A and B). This percentage reduced by one-half on day 1 of co-culture (43.78%; SE, 3.03; P=0.000264 vs. cells cultured alone; Fig. 3C). Despite this population size increasing over time (day 3, 59.50%; SE, 2.41; P=0.000449 vs. cells cultured alone; day 7, 78.36%; SE, 2.19; P=0.015462 vs. cells cultured alone; Fig. 3D and E, respectively), the original population size detected in ECs cultured alone was not completely reestablished at day 10 of co-culture (80.73%; SE, 2.06; P=0.000622 vs. cells cultured alone; Fig. 3F).

Phenotypic profile of co-cultured ECs over time

According to the variations detected within the populations of ECs cultured alone or co-cultured, the stromal endometrial cell marker CD13 (42,43) and the endometrial mesenchymal stem cell (eMSC) marker CD146 (4446) were selected to further characterize these populations. CD13 expression was not significantly different within co-cultured or individually cultured populations (Fig. 4).

The expression of eMSC marker CD146 varied significantly over time (Fig. 5). Population one had negligible expression of CD146, with the mean expression at all time points in the co-cultured group of 1.21% (SE, 0.25). In population two, an increase in CD146 expression was observed between days 1 and 7 of co-culture (1st vs. 3rd days, P=0.037; 3rd vs. 7th days, P=0.017), with a slight decrease in expression between days 7 and 10 of co-culture (P=0.071). The expression of CD146 in population 2 at 1st day of co-culture was more similar to endometriotic cells (P=0.102010) compared with healthy ECs (P=0.051579) (data not shown). These results demonstrated that population two was predominantly composed of eMSCs, and that there was an increase in these cells when healthy and endometriotic cells were co-cultured.

Discussion

The first and most acceptable theory for endometriosis pathogenesis was proposed almost a century ago (47), yet the pathophysiology of this disease remains unclear. As a hormone-dependent and chronic inflammatory disease (1,2,8), cytokines and soluble factors appear to serve an important role in its pathophysiology. The inflammatory content of the peritoneal fluid and serum, in addition to the eutopic and ectopic endometrium has been extensively studied (6,11,12,6). However, there are a number of questions which remain unanswered.

The increased levels of inflammatory cytokines in the peritoneal fluid, serum and endometrium of women with end ometriosis has been reported in numerous studies (18,23,28,18). The principal identified factors include TNF-α, IL-6, IL-8, IL-10, VEGF and MCP-1 (11,18,2124,4951). In the present study, increased levels of IL-6 and IL-8 were observed in endometriotic cells cultured alone and in co-culture with healthy ECs, compared with individually cultured healthy ECs. The levels of IL-1β were additionally increased in the co-culture at day 10.

Sikora et al (18) proposed that IL-8 is important for progression of endometriosis. IL-8 increases the adhesion of endometrial stroma to extracellular matrix proteins, in addition to metalloproteinase expression and cell proliferation (2628). The surgical excision of endometrial lesions leads to a decrease in the peritoneal levels of IL-8 (38). In the present study, increased secretion of IL-8 by endometriotic and co-cultured cells was detected. A previous study indicated that peritoneal immune cells, particularly macrophages, are the main source of IL-8 (18). The results of the present study indicated that ECs additionally contribute to IL-8 secretion. Furthermore, IL-8 expression was moderately correlated with cell number and IL-6 expression, suggesting that IL-8 secretion may be independent of cell proliferation and IL-6 levels.

IL-6 is secreted by the endometrium and implants, and is involved in diverse aspects of reproductive physiology, including ovarian steroid production, foliculogenesis and early embryonic implantation (28,49,52,53). IL-6 is associated with the increased expression of ICAM-1 by macrophages in patients with endometriosis (29,30). The present study demonstrated that the secretion of IL-6 by endometriotic cells was higher compared with healthy or co-cultured ECs. The increased levels of IL-6 were strongly correlated with cell numbers, indicating that the increased proliferative rate may be due to IL-6 signaling. The increased cell number also increases the IL-6 secretion. It has been well established that endometriotic stromal cells have a higher proliferative rate compared with healthy endometrial stromal cells (54,55). This increase in proliferation rate may be mediated by IL-6.

The levels of IL-6 may be associated with IL-1β secretion (20). The results of the present study revealed the presence of IL-1β at day 10 of co-culture only, with no significant alterations in the expression of IL-1β in individually cultured healthy or endometriotic cells. Furthermore, there was no statistically significant correlation between IL-6 and IL-1β expression. The secretion of IL-1β by the primary co-cultured ECs may be associated with long-term co-culture, which may indicate a switch from an acute to chronic inflammatory response in the endometrial cells (21). The proliferative effects of IL-1β have been reported in endometriotic cells (13). Furthermore, an increase in IL-1B expression occurs in the co-culture of endometrial primary cells and MSCs (56).

The endometrium contains MSCs that are involved in the cyclical regeneration of this tissue (5759). The role of stem cells in the pathogenesis of endometriosis has been reported (37,60,61). The most widely accepted theory for endometriosis pathogenesis was proposed by Sampson (47) and indicates that the implants establish from cells undergoing retrograde menstruation; this theory alone is unable to explain all clinical presentations of the disease. The combined theories of Sampson (47) and the involvement of stem cells provide a better explanation for endometriosis physiopathology. The dysregulation of eMSCs has been proposed as a key mechanism of endometriosis, in concordance with the retrograde menstruation theory (62).

The mesenchymal stem cell marker CD146 (46) was used in the present study in order to identify the presence of MSCs in the pool of primary cells, and to report the variations in the MSC population during co-culture. An increase in the number of eMSCs detected by CD146 expression was observed in the co-culture system. These alterations in CD146 expression over time, together with the alterations in cytokine secretion, may indicate the involvement of MSCs in cytokine secretion. A pool of primary cells at a low passage number were selected, as this contained various cell populations and was more similar to the composition of the eutopic endometrium. In previous work (56), the co-culture of umbilical cord blood MSCs with primary endometrial cells resulted in increased IL-1β expression. Furthermore, co-culture eliminated the effect of p27 gene therapy on endometriotic stromal cells. These observations, together with the identification and characterization of endometrial MSCs (37,45,58,37), have provided an insight into the involvement of MSCs in endometriosis pathogenesis. In the present study, the co-cultivation of a pool of healthy and diseased primary cells, containing endometrial MSCs, led to the secretion of cytokines that may be directly involved in the pathogenesis of endometriosis. This supports the theory that eMSCs have an important role in the secretion of cytokines and endometriosis.

MSCs express CD13 (44,46), which is additionally expressed by stromal endometrial cells (42,43). CD13 is an N-aminopeptidase involved in the inactivation of IL-8 (64). Conflictingly, increased levels of CD13 expression due to cell-to-cell contact may be associated with IL-8 inactivation resistance (65) and the inhibition of apoptosis mediated by IL-8 (66). In the present study, CD13 expression was not significantly different between the co-cultured and individually cultured cells, indicating no physiological implications. Therefore, CD13 expression in the present study may be indicative of fibroblastoid cells.

Furthermore, the present study observed that a mixed cell population derived from the endometrium exhibited a secretory profile similar to that of endometriotic cells, even in the presence of healthy ECs. The results obtained demonstrated that the communication between endometriotic and healthy cells results in a secretory profile similar to what is reported in the peritoneal fluid of women with endometriosis (24,28,31,52). The increased secretion of IL-6 and IL-8 is additionally associated with the progesterone resistance observed in endometriosis (32,34). The results of the present study support this finding, as the endometriotic cells secreted higher levels of IL-6 and IL-8 compared with the healthy cells. Additionally, the findings of the present study were consistent with the retrograde menstruation theory and clarified certain aspects of the disrupted peritoneal environment of women with endometriosis. For example, despite the presence of healthy endometrial cells in retrograde menstruation, the presence of endometriotic cells may be decisive in the secretory profile of the cell pool (18,21,37). The endometrial cells regurgitated into peritoneal cavity may be responsible for the induction of the aberrant cytokine profile observed in the peritoneal fluid and eutopic endometrium of women with endometriosis (18,19,21,18). The variations observed in the CD146+ population support the putative role of eMSCs in the pathogenesis of endometriosis.

The secretion alterations observed in the individually cultured ECs and the co-culture indicate a critical role for ectopic endometrial cells in the initiation of peritoneal environment disruption, which is in accordance with a recently published report (38). Thus, the cytokine profile observed may not only be generated by molecules secreted from immune cells. The main limitation of the present study is that, as an in vitro study, the effects of EC-secreted cytokines on immune cells present in the peritoneum of women with endometriosis was not demonstrated. Studies which isolate each population of cells observed in the present study may clarify the cell type responsible for the secretion of the cytokines or reveal the association between healthy and disease cell contact.

In accordance to previous research (18,19,21,18), the findings of the present study suggest that the soluble factors secreted by endometriotic cells may have an important role in the disruption of the cell cycle and in the establishment of the peritoneal environment of women with endometriosis. The endometriotic cell pool may be responsible for the establishment of an inflammatory peritoneal environment favorable to the initiation and progression of endometrial cell adhesion and clustering.

Acknowledgements

We thank Colsan Blood Bank (São Paulo, Brazil) for all the technical support.

Funding

The present study was supported by the São Paulo Research Foundation (grant no. 2011/14683-7) and the Brazilian National Council of Technological and Scientific Development (grant no. 480303/2013-4).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

ALI, GAG, MJBCG and ES provided major contributions to the conception and design of the present study. ALI, RMP, AK and GK conducted the collection of samples, experimentation and acquisition of data. ALI and ES were involved in analysis and interpretation of data. All authors were involved in manuscript drafting and critical discussion, as well as in the final approval of the version to be published.

Ethics approval and consent to participate

The protocol of the present study was approved by the Federal University of São Paulo ethical committee (São Paulo, Brazil). Written informed consent was obtained from all patients.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ulukus M, Cakmak H and Arici A: The Role of Endometrium in Endometriosis. J Soc Gynecol Investig. 13:467–476. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Falconer H, D'Hooghe T and Fried G: Endometriosis and genetic polymorphisms. Obstet Gynecol Surv. 62:616–628. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Eskenazi B and Warner ML: Epidemiology of endometriosis. Obstet Gynecol Clin North Am. 24:235–258. 1997. View Article : Google Scholar : PubMed/NCBI

4 

Vinatier D, Cosson M and Dufour P: Is endometriosis an endometrial disease? Eur J Obstet Gynecol Reprod Biol. 91:113–125. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Bulun SE: Endometriosis. N Engl J Med. 360:268–279. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Burney RO and Giudice LC: Pathogenesis and pathophysiology of endometriosis. Fertil Steril. 98:511–519. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Giudice LC and Kao LC: Endometriosis. Lancet. 364:1789–1799. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Berkley KJ, Rapkin AJ and Papka RE: The Pains of Endometriosis. Science. 308:1587–1589. 2005. View Article : Google Scholar : PubMed/NCBI

9 

de Marqui Trovó AB: Genetic polymorphisms and endometriosis: Contribution of genes that regulate vascular function and tissue remodeling. Rev Assoc Med Bras. 58:620–32. 2012.(In English, Portuguese). PubMed/NCBI

10 

Kobayashi H, Yamada Y, Morioka S, Niiro E, Shigemitsu A and Ito F: Mechanism of pain generation for endometriosis-associated pelvic pain. Arch Gynecol Obstet. 289:13–21. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Ilie I and Ilie R: Cytokines and endometriosis-the role of immunological alterations. Biotechnol Mol Biol Nanomedicine. 1:8–19. 2013.

12 

Gazvani R and Templeton A: Peritoneal environment, cytokines and angiogenesis in the pathophysiology of endometriosis. Reproduction. 123:217–26. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Lebovic DI, Baldocchi RA, Mueller MD and Taylor RN: Altered expression of a cell-cycle suppressor gene, Tob-1, in endometriotic cells by cDNA array analyses. Fertil Steril. 78:849–854. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Bersinger NA, Günthert AR, McKinnon B, Johann S and Mueller MD: Dose-response effect of interleukin (IL)-1β, tumour necrosis factor (TNF)-α, and interferon-γ on the in vitro production of epithelial neutrophil activating peptide-78 (ENA-78), IL-8, and IL-6 by human endometrial stromal cells. Arch Gynecol Obstet. 283:1291–1296. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Yoshino O, Izumi G, Shi J, Osuga Y, Hirota Y, Hirata T, Harada M, Nishii O, Koga K and Taketani Y: Activin-A is induced by interleukin-1β and tumor necrosis factor-α and enhances the mRNA expression of interleukin-6 and protease-activated receptor-2 and proliferation of stromal cells from endometrioma. Fertil Steril. 96:118–121. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Bilotas M, Meresman G, Buquet R, Sueldo C and Barañao RI: Effect of vascular endothelial growth factor and interleukin-1beta on apoptosis in endometrial cell cultures from patients with endometriosis and controls. J Reprod Immunol. 84:193–198. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Vetvicka V, Laganà AS, Salmeri FM, Triolo O, Palmara VI, Vitale SG, Sofo V and Králíčková M: Regulation of apoptotic pathways during endometriosis: From the molecular basis to the future perspectives. Arch Gynecol Obstet. 294:897–904. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Sikora J, Smycz-Kubańska M, Mielczarek-Palacz A and Kondera-Anasz Z: Abnormal peritoneal regulation of chemokine activation-The role of IL-8 in pathogenesis of endometriosis. Am J Reprod Immunol. 77:2017. View Article : Google Scholar : PubMed/NCBI

19 

Gonçalves GA, Camargo-Kosugi CM, Bonetti TC, Invitti AL, Girão MJ, Silva ID and Schor E: p27kip1 overexpression regulates VEGF expression, cell proliferation and apoptosis in cell culture from eutopic endometrium of women with endometriosis. Apoptosis. 20:327–335. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Kyama CM, Overbergh L, Mihalyi A, Meuleman C, Mwenda JM, Mathieu C and D'Hooghe TM: Endometrial and peritoneal expression of aromatase, cytokines, and adhesion factors in women with endometriosis. Fertil Steril. 89:301–310. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Sikora J, Mielczarek-Palacz A and Kondera-Anasz Z: Association of the precursor of interleukin-1β and peritoneal inflammation-role in pathogenesis of endometriosis. J Clin Lab Anal. 30:831–837. 2016. View Article : Google Scholar : PubMed/NCBI

22 

May CD, Sphyris N, Evans KW, Werden SJ, Guo W and Mani SA: Epithelial-mesenchymal transition and cancer stem cells: A dangerously dynamic duo in breast cancer progression. Breast Cancer Res. 13:2022011. View Article : Google Scholar : PubMed/NCBI

23 

Fan YY, Chen HY, Chen W, Liu YN, Fu Y and Wang LN: Expression of inflammatory cytokines in serum and peritoneal fluid from patients with different stages of endometriosis. Gynecol Endocrinol. 1–6. 2018.(Epub ahead of Print).

24 

Jørgensen H, Hill AS, Beste MT, Kumar MP, Chiswick E, Fedorcsak P, Isaacson KB, Lauffenburger DA, Griffith LG and Qvigstad E: Peritoneal fluid cytokines related to endometriosis in patients evaluated for infertility. Fertil Steril. 107:1191–1199.e2. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Sakamoto Y, Harada T, Horie S, Iba Y, Taniguchi F, Yoshida S, Iwabe T and Terakawa N: Tumor necrosis factor-alpha-induced interleukin-8 (IL-8) expression in endometriotic stromal cells, probably through nuclear factor-kappa B activation: Gonadotropin-releasing hormone agonist treatment reduced IL-8 expression. J Clin Endocrinol Metab. 88:730–735. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Garcia-Velasco JA and Arici A: Interleukin-8 stimulates the adhesion of endometrial stromal cells to fibronectin. Fertil Steril. 72:336–340. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Arici A, Seli E, Zeyneloglu HB, Senturk LM, Oral E and Olive DL: Interleukin-8 induces proliferation of endometrial stromal cells: A potential autocrine growth factor. J Clin Endocrinol Metab. 83:1201–1205. 1998. View Article : Google Scholar : PubMed/NCBI

28 

Kalu E, Sumar N, Giannopoulos T, Patel P, Croucher C, Sherriff E and Bansal A: Cytokine profiles in serum and peritoneal fluid from infertile women with and without endometriosis. J Obstet Gynaecol Res. 33:490–495. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Herington JL, Bruner-Tran KL, Lucas JA and Osteen KG: Immune interactions in endometriosis. Expert Rev Clin Immunol. 7:611–626. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Young VJ, Brown JK, Saunders PT and Horne AW: The role of the peritoneum in the pathogenesis of endometriosis. Hum Reprod Update. 19:558–569. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Barcz E, Milewski Ł, Dziunycz P, Kamiński P, Płoski R and Malejczyk J: Peritoneal cytokines and adhesion formation in endometriosis: An inverse association with vascular endothelial growth factor concentration. Fertil Steril. 97:1380–1386.e1. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Young SL and Lessey BA: Progesterone function in human endometrium: Clinical perspectives. Semin Reprod Med. 28:5–16. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Barragan F, Irwin JC, Balayan S, Erikson DW, Chen JC, Houshdaran S, Piltonen TT, Spitzer TL, George A, Rabban JT, et al: Human endometrial fibroblasts derived from mesenchymal progenitors inherit progesterone resistance and acquire an inflammatory phenotype in the endometrial niche in endometriosis. Biol Reprod. 94:1182016. View Article : Google Scholar : PubMed/NCBI

34 

Lessey BA and Young SL: Homeostasis imbalance in the endometrium of women with implantation defects: The role of estrogen and progesterone. Semin Reprod Med. 32:365–375. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Wing LY, Chuang PC, Wu MH, Chen HM and Tsai SJ: Expression and mitogenic effect of fibroblast growth factor-9 in human endometriotic implant is regulated by aberrant production of estrogen. J Clin Endocrinol Metab. 88:5547–5554. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Wu MH, Lu CW, Chuang PC and Tsai SJ: Prostaglandin E2: The master of endometriosis? Exp Biol Med (Maywood). 235:668–677. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Gargett CE, Schwab KE, Brosens JJ, Puttemans P, Benagiano G and Brosens I: Potential role of endometrial stem/progenitor cells in the pathogenesis of early-onset endometriosis. Mol Hum Reprod. 20:591–598. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Monsanto SP, Edwards AK, Zhou J, Nagarkatti P, Nagarkatti M, Young SL, Lessey BA and Tayade C: Surgical removal of endometriotic lesions alters local and systemic proinflammatory cytokines in endometriosis patients. Fertil Steril. 105:968–977.e5. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Moen MH and Halvorsen TB: Histologic confirmation of endometriosis in different peritoneal lesions. Acta Obstet Gynecol Scand. 71:337–342. 1992. View Article : Google Scholar : PubMed/NCBI

40 

Revised American Society for Reproductive Medicine classification of endometriosis: 1996. Fertil Steril. 67:817–821. 1997. View Article : Google Scholar : PubMed/NCBI

41 

Vieira S: Introdução à Bioestatística. 4th edition. Elsevier Ltd; São Paulo: 2008

42 

Yoshimoto Kato M, Kato K, Adachi S, Yamayoshi A, Arima T, Asanoma K, Kyo S, Nakahata T and Wake NK: Characterisation of side population cells in human normal endometrium. Hum Reprod. 22:1212–1223. 2007.

43 

Seli E, Senturk LM, Bahtiyar OM, Kayisli UA and Arici A: Expression of aminopeptidase N in human endometrium and regulation of its activity by estrogen. Fertil Steril. 75:1172–1176. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Schwab KE and Gargett CE: Co-expression of two perivascular cell markers isolates mesenchymal stem-like cells from human endometrium. Hum Reprod. 22:2903–2911. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Schwab KE, Hutchinson P and Gargett CE: Identification of surface markers for prospective isolation of human endometrial stromal colony-forming cells. Hum Reprod. 23:934–943. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Gargett CE, Schwab KE, Zillwood RM, Nguyen HP and Wu D: Isolation and culture of epithelial progenitors and mesenchymal stem cells from human endometrium. Biol Reprod. 80:1136–1145. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Sampson JA: Peritoneal endometriosis due to the menstrual dissemination of endometrial tissue into the peritoneal cavity. Am J Obstet Gynecol. 14:422–469. 1927. View Article : Google Scholar

48 

Milewski Ł, Dziunycz P, Barcz E, Radomski D, Roszkowski PI, Korczak-Kowalska G, Kamiński P and Malejczyk J: Increased levels of human neutrophil peptides 1, 2, and 3 in peritoneal fluid of patients with endometriosis: Association with neutrophils, T cells and IL-8. J Reprod Immunol. 91:64–70. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Jacobs AL, Sehgal PB, Julian J and Carson DD: Secretion and hormonal regulation of interleukin-6 production by mouse uterine stromal and polarized epithelial cells cultured in vitro. Endocrinology. 131:1037–1346. 1992. View Article : Google Scholar : PubMed/NCBI

50 

Szyllo K, Tchorzewski H, Banasik M, Glowacka E, Lewkowicz P and Kamer-Bartosinska A: The involvement of T lymphocytes in the pathogenesis of endometriotic tissues overgrowth in women with endometriosis. Mediators Inflamm. 12:131–138. 2003. View Article : Google Scholar : PubMed/NCBI

51 

Kang YJ, Jeung IC, Park A, Park YJ, Jung H, Kim TD, Lee HG, Choi I and Yoon SR: An increased level of IL-6 suppresses NK cell activity in peritoneal fluid of patients with endometriosis via regulation of SHP-2 expression. Hum Reprod. 29:2176–2189. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Harada T, Yoshioka H, Yoshida S, Iwabe T, Onohara Y, Tanikawa M and Terakawa N: Increased interleukin-6 levels in peritoneal fluid of infertile patients with active endometriosis. Am J Obstet Gynecol. 176:593–597. 1997. View Article : Google Scholar : PubMed/NCBI

53 

Bedaiwy MA, Falcone T, Sharma RK, Goldberg JM, Attaran M, Nelson DR and Agarwal A: Prediction of endometriosis with serum and peritoneal fluid markers: A prospective controlled trial. Hum Reprod. 17:426–431. 2002. View Article : Google Scholar : PubMed/NCBI

54 

D'Amora P, Maciel TT, Tambellini R, Mori MA, Pesquero JB, Sato H, Girão MJ, da Silva Guerreiro ID and Schor E: Disrupted cell cycle control in cultured endometrial cells from patients with endometriosis harboring the progesterone receptor polymorphism PROGINS. Am J Pathol. 175:215–224. 2009. View Article : Google Scholar : PubMed/NCBI

55 

Li MQ, Luo XZ, Meng YH, Mei J, Zhu XY, Jin LP and Li DJ: CXCL8 enhances proliferation and growth and reduces apoptosis in endometrial stromal cells in an autocrine manner via a CXCR1-triggered PTEN/AKT signal pathway. Hum Reprod. 27:2107–2116. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Gonçalves GA, Invitti AL, Parreira RM, Kopelman A, Schor E and Girão MJ: p27kip1 overexpression regulates IL-1β in the microenvironment of stem cells and eutopic endometriosis co-cultures. Cytokine. 89:229–234. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Prianishnikov VA: On the concept of stem cell and a model of functional-morphological structure of the endometrium. Contraception. 18:213–223. 1978. View Article : Google Scholar : PubMed/NCBI

58 

Gargett CE, Chan RW and Schwab KE: Endometrial stem cells. Curr Opin Obstet Gynecol. 19:377–383. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Sasson IE and Taylor HS: Stem cells and the pathogenesis of endometriosis. Ann N Y Acad Sci. 1127:106–115. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Gargett BE and Chan RW: Endometrial stem/progenitor cells and proliferative disorders of the endometrium. Minerva Ginecol. 58:511–526. 2006.PubMed/NCBI

61 

Gargett CE and Gurung S: Endometrial mesenchymal stem/stromal cells, their fibroblast progeny in endometriosis, and more1. Biol Reprod. 94:1292016. View Article : Google Scholar : PubMed/NCBI

62 

Hufnagel D, Li F, Cosar E, Krikun G and Taylor H: The role of stem cells in the etiology and pathophysiology of endometriosis. Semin Reprod Med. 33:333–340. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Gargett CE and Masuda H: Adult stem cells in the endometrium. Mol Hum Reprod. 16:818–834. 2010. View Article : Google Scholar : PubMed/NCBI

64 

Kanayama N, Kajiwara Y, Goto J, el Maradny E, Maehara K, Andou K and Terao T: Inactivation of interleukin-8 by aminopeptidase N (CD13). J Leukoc Biol. 57:129–134. 1995. View Article : Google Scholar : PubMed/NCBI

65 

Kehlen A, Egbert I, Thiele K, Fischer K, Riemann D and Langner J: Increased expression of interleukin-8 and aminopeptidase N by cell-cell contact: Interleukin-8 is resistant to degradation by aminopeptidase N/CD13. Eur Cytokine Netw. 12:316–324. 2001.PubMed/NCBI

66 

Mishima Y, Matsumoto-Mishima Y, Terui Y, Katsuyama M, Yamada M, Mori M, Ishizaka Y, Ikeda K, Watanabe J, Mizunuma N, et al: Leukemic cell-surface CD13/aminopeptidase N and resistance to apoptosis mediated by endothelial cells. J Natl Cancer Inst. 94:1020–1028. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 18 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luckow Invitti A, Schor E, Martins Parreira R, Kopelman A, Kamergorodsky G, Gonçalves GA and Batista Castello Girão MJ: Inflammatory cytokine profile of co‑cultivated primary cells from the endometrium of women with and without endometriosis. Mol Med Rep 18: 1287-1296, 2018
APA
Luckow Invitti, A., Schor, E., Martins Parreira, R., Kopelman, A., Kamergorodsky, G., Gonçalves, G.A., & Batista Castello Girão, M.J. (2018). Inflammatory cytokine profile of co‑cultivated primary cells from the endometrium of women with and without endometriosis. Molecular Medicine Reports, 18, 1287-1296. https://doi.org/10.3892/mmr.2018.9137
MLA
Luckow Invitti, A., Schor, E., Martins Parreira, R., Kopelman, A., Kamergorodsky, G., Gonçalves, G. A., Batista Castello Girão, M. J."Inflammatory cytokine profile of co‑cultivated primary cells from the endometrium of women with and without endometriosis". Molecular Medicine Reports 18.2 (2018): 1287-1296.
Chicago
Luckow Invitti, A., Schor, E., Martins Parreira, R., Kopelman, A., Kamergorodsky, G., Gonçalves, G. A., Batista Castello Girão, M. J."Inflammatory cytokine profile of co‑cultivated primary cells from the endometrium of women with and without endometriosis". Molecular Medicine Reports 18, no. 2 (2018): 1287-1296. https://doi.org/10.3892/mmr.2018.9137