Abstract
Endometriosis is a prevalent chronic disease that affects approximately 6% to 10% of reproductive-aged women. Although numerous researchers have endeavored to explore the etiology of endometriosis over a century, its etiology still remains an enigma. The exploration of pathophysiologic mechanism and novel therapy for endometriosis depends on ideal endometriotic models. In the previous decade, various endometriotic models have been established; therefore, we made a conclusion for available information on these models. This review summarized the common experimental models used in endometriotic studies, including their origins, characteristics, applications, and limitations. Endometriotic models played an important role in studying etiologies and novel treatments of endometriosis during the last decades. Among them, animal models and endometriotic cell lines were viewed as most common studying tools to explore the intrinsic entities of endometriosis. In addition, endometrial organoid also emerged and was regarded as an ideal studying tool for endometriosis research. Different research models collectively complement each other to advance the endometriosis research. The successful establishment of endometrial organoids means that organoids are expected to become an ideal model for studying endometriosis in the future.
Keywords: Endometriosis, Endometriotic studying models, Endometriotic cell lines, Animal models, Endometrial organoids
Introduction
As a prevalent disease, endometriosis affects approximately 6% to 10% of all women during their reproductive age.[1] The incidence of endometriosis ranges within 40% to 60% in women with dysmenorrhea and within 20% to 30% in women with subfertility.[2,3] Endometriosis is characterized by the presence of endometrioid epithelial and stromal cells outside the uterus. Endometriosis is mostly found in the pelvic cavity, and mainly consists of three categories: peritoneal, ovarian, and deep infiltrating endometriosis (DIE).[4,5] Patients often experience pelvic pain, dysmenorrhea, deep dyspareunia, ovary cyst and infertility, and even face a higher risk of epithelial ovarian cancer.[4,6,7] Present treatments contain pain medication, hormonal intervention, and surgery.[7]
Despite these numerous studies, the etiologies of endometriosis remain indistinct, which depends on ideal studying models. Among these, animal models and endometriotic cell lines are viewed as the most common tools to support the research of endometriosis. In addition, the basis on organoid cultures that concern the liver and gut, endometrial organoid (EO) has also emerged, and has been regarded as a reliable model for endometriosis research. Therefore, the present review summarizes the experimental models that have been used in endometriotic studies, and it was considered that organoid culture could be an ideal model complementary to others for endometriosis study in the future.
Published literatures were searched from the PubMed, Embase, and Web of Science databases with the following relative terms: “endometriosis cell line∗,” “(endometriosis epithelial cell line∗[Title/Abstract]) OR endometriosis stromal cell line∗[Title/Abstract]),” “(endometriosis) AND (stem cell),” “organoid culture,” “(endometrium) AND (organoid culture),” and “(endometriosis) AND (animal models).” The search was performed until December 2019. In addition, relevant reviews and the reference lists of all the included articles were analyzed to search for related articles.
In Vitro Cell Models
The characteristics and potential applications of endometriotic cell lines are respectively presented in Tables 1 and 2. In addition, the details of the in vitro cell models were introduced in each part.
Table 1.
The cell lines used in endometriosis studies.

Table 2.
Potential applications of endometriotic cell lines for preclinical research.

Characteristics of endometriotic primary cells
Gaetje et al reported that the E-cadherin negative epithelial cell of endometriotic primary cells is invasive, and could be distinguished from fibroblasts and stromal cells by its expression of cytokeratin.[18] Hence, this cell type, cytokeratin+/E-cadherin−, might play an important role in the development and invasion of endometriosis.[19] Primary cells can better characterize the disease, but have a limited life span. This lay a foundation for the subsequent establishment of endometriosis cell lines.
Endometriotic epithelial cell line 145T
Endometriotic epithelial cell line (EEC) 145T was established, and the detailed procedure was previously described.[18,19] Derived from peritoneal biopsies and transformed Simian virus 40 (SV40) T antigen,[20] EEC145T is invasive, cytokeratin+/E-cadherin−, and is expressed both in the estrogen receptor (ER) and progesterone receptor (PR) by immunocytochemical analyses, with a life span approximately 35 passages. At about passage 25, EEC145T begins to lose the expression of cytokeratin, and the invasive features and expression of fibroblast growth factor activating gene 1 (Frag-1) mRNA, but maintains its proliferative potential.[20] Thus, EEC145T can retain these initial characteristics for as long as 25 passages. Furthermore, the addition of 10% (v/v) peritoneal fluid considerably enhanced the invasive capacity of EEC145, while the removal of steroids and growth factors in peritoneal fluid or the treatment at 95°C did not change the invasion-promoting activity.[20] As a studying tool, EEC145T was once used to verify that ovarian cancer antigen CA125 influences the cell adhesion in vitro.[21]
EEC10Z, EEC11Z, EEC12Z, EEC49Z
Zeitvogel et al established a number of endometriotic cell lines derived from peritoneal endometriotic biopsies.[22] As immortalized cell lines using the SV40 T antigen, these endometriotic cell lines included two categories: one category exhibited stromal cell features, while the other category exhibited epithelial-like features. However, both of these were E-cadherin negative. Among these, cell lines 10Z, 12Z, 39Z, and 50Z escaped the crisis, and became immortal. Furthermore, cell lines 10Z, 11Z, 12Z, and 49Z, and the previous cell line EEC145T were selected for further investigation [Table 1]. Although E-cadherin was negative, N-cadherin was expressed in endometriotic cell lines, which is similar to metastatic EJ28 cells.[22] This coincided with previous studies, indicating that N-cadherin might be associated with invasion and migration.[23] Notably, Zeitvogel et al provided many useful study tools for endometriosis research, especially EEC12Z, which has been most frequently used. More than 40 studies have used this as a research tool. Although there was no more introduction about endometriotic stromal cell (ESC) 22B, ESC22B has been widely employed as a study tool, especially in cooperation with EEC12Z.[8,10,14] In addition, some cell lines, such as 11Z, 12Z, 49Z, 108Z, and 22B, have been further examined by Banu et al.[24] They focused on exploring gene expression profiles and the functional characterization of these cell lines, such as the mRNA expression of relevant cytokines and cell cycle regulation.[24]
EMosis-CC/TERT1 and EMosis-CC/TERT2
Due to the risk of carcinogenesis in endometrioma,[25] it is vital to study the ovarian EEC line, to explore carcinogenesis mechanism and treatment. However, ectopic ovarian epithelial cell lines are more difficult to culture, when compared to ectopic stromal cell lines,[26] and premature senescence and telomere-dependent senescence were the two paramount limitations.[27] To solve this problem, Bono et al attempted be different combinations of transfection genes, and produced five independent cell populations from two patients by co-transfection of at least three genes.[28] These cells could grow for over 100 population doubling, maintaining the original morphology and negative senescence-associated β-gal staining.[28] The outcomes indicated that the co-expression of cyclinD1 and cdk4 could overcome the premature senescence of EECs, and that combining these with hTERT is sufficient for the immortal phenotypes, without the additional inactivation of p53. Then, they produced the EEC lines, parental cells, EMosis-CC/TERT1, and EMosis-CC/TERT2.[28] Unfortunately, the EMosis-CC/TERT2 was contaminated with few interstitial cells. For the response to steroid hormone, the immortal epithelial cells exhibited a cell response to progestin, but there was no response to estrogen (E2). These managed to overexpress ERα in EMOsis-CC/TERT1 cells via the lentiviral introduction of ERα cDNA and obtained the ERα-over-expressing cell line EMOsis-CC/TERT1/ER. The sufficient expression of ERα in EMOsis-CC/TERT1/ER was confirmed by western blot, and the growth of EMOsis-CC/TERT1/ER cells was markedly activated by the addition of E2.[28] Finally, these successfully generated immortal EECs from ovarian endometrioma that still had the property of estrogen or progestin response and did not have tumorigenicity: EMOsis-CC/TERT1/ER cell line. In general, this cell line has been a useful tool for endometriosis research, especially for the carcinogenesis of ovarian endometrioma. In the introduction or knockdown of candidate genetic factors, this cell line enabled the identification of genetic factors required for transformation. For example, Mita et al used EMosis-CC/TERT1 and its deuterogenic cell lines to explore the pharmacologic mechanism of dienogest.[29]
EEC16 and EEC16-TERT
Brueggmann et al established an EEC16 from ovarian surface endometriosis lesions.[30] They cultured EEC16, EEC12Z, and normal ovarian surface epithelial cells (OSECs) in vitro three-dimensional (3D) culture models and examined morphologic and molecular characteristics. EEC16 displayed an epithelial morphology with mesenchymal factors: cytokeratin+/vimentin+, but E-cadherin−/N-cadherin−/P-cadherin−/ERα−.[30] After performing RNA-sequencing to compare the transcriptome between primary EEC16 and OSEC, Brueggmann et al found 1780 significantly differentially expressed genes. Intriguingly, EEC16 more closely resembled peritoneal lesions than cystic endometriomas within the ovarian cortex on the histologic examination. Compared with the two-dimensional model, the 3D culture model could better represent the characteristics of the endometriosis expression profile.[30]
Based on EEC16, Lawrenson et al established a novel TERT (human telomerase) immortalized cell line (EEC16-TERT).[31] The lifespan of EEC16 in vitro was increased following the transduction with either lentiviral-TERT (EEC16-TERT-L), or retroviral-TERT (EEC16-TERT-R), when compared to both control cells transduced with GFP (EEC16-GFP) and primary EEC16, from 60 to 70 days extended to over 200 days. In addition, EEC16-TERT retained the normal karyotype, and did not appear with tumorigenicity in vivo.[31]
Primary and TERT-expressing EEC16 shared similar morphologies, but had differentially expressed epithelial and mesenchymal markers: the primary EEC16 cell was cytokeratin+/vimentin+/E-cadherin−,[30] while the EEC16-TERT cell was cytokeratin−/vimentin+, suggesting the occurrence of epithelial-mesenchymal-transition (EMT).[31] EMT is a key process in the pathogenesis of endometriosis[32] which does not occur in TERT-immortalized OSECs,[33] suggesting that this phenomenon is a feature of EEC16 and/or other endometriosis-derived cell lines. Furthermore, compared to primary EEC16, EEC16-TERT cultures exhibited a lower expression of tumor suppressor genes associated with clear cell ovarian cancer (ARID1A, MLH1, and MSH2).[31] Lawrenson et al hypothesized that Src activation could be a driver of endometriosis-associated ovarian cancer (EAOC). If the Src signature is differentially activated in the eutopic endometrium of women at highest risk of developing EAOC, endometrial biopsy might represent a non-invasive screening tool to detect early stage EAOC.[31] Therefore, EEC16-TERT is a potential model to deeply study the pathogenesis and identify the novel therapy for endometriosis, especially for EOAC.
In fact, as early as 2012, Boccellino et al reported the introduction of hTERT to construct EEC lines and stromal cell lines derived from deep endometriotic tissues. In addition to the absence of chromosomal abnormalities and long-term expansion, the cell line also maintains the natural characteristics of endometrial cells from the perspective of the phenotype and functional expression of estrogen and PRs.[34] In their study, for immortalized cell lines, epithelial cells expressed cytokeratin 7 and stromal cells expressed CD10. However, it was uncertain whether EMT would occur in later passages.
The ESC line Hs832cT (CRL-7566)
CRL-7566 was established by American Type Culture Collection (ATCC; Manassas, VA, USA). This cell line, which was derived from an ovarian cyst wall from a patient with endometriosis, has been partially characterized as stromal (vimentin+/cytokeratin−/E-cadherin−). This cell line had been broadly used as a research tool.[9,12,16]
Endometrial stromal cell line St-T1b
Samalecos et al established an ESC line St-T1b, which was immortalized by introducing hTERT into primary cells derived from the proliferative endometrium of patients with tubal disorder.[35] St-T1b displays the phenotype of vimentin+/CD90+/CK7− by immunocytochemistry.[35] By reverse transcription polymerase chain reaction, PR, and ERα were readily detectable in St-T1b cells, and the ERβ transcripts were below the limit of detection.[35] However, neither PR-B nor PR-A was detectable by western blotting in St-T1b cells, and the combination of progestin with E2 failed to induce morphologic changes associated with decidualization in St-T1b cells.[35] Although cells did not respond to progestin alone, Samalecos et al demonstrated that cyclic AMP (cAMP)-induced St-T1b cells respond to progestin, and the combination of cAMP with synthetic progestin MPA promoted the decidualization better, when compared to P4.[35] However, these phenomena would decline in later passages. In addition, when compared to that in vivo, E2 did not induce PR protein in cells, and no induction was obtained with steroids alone. Although St-T1b was originally established to study the decidualization, this could be used as a contrast to the endometriotic cell line while studying the etiology of endometriosis, such as the role of micro-200b in endometriosis.[13]
CRL-4003, which is a hTERT-immortalized human ESC line (T-HESC) established by the ATCC (CRL-4003), has also been frequently used as contrast in endometriosis research.[36,37] Furthermore, Kyo et al, Krikun et al, Barbier et al, and Chapdelaine et al also established some endometrial cell lines, which might be useful in endometriosis research.[38–41]
Epithelial progenitors and mesenchymal stem cells obtained from human endometrium
More than 90% reproductive women experienced retrograde menstruation, but merely 6% to 10% suffered from endometriosis.[42] Some experts hypothesized that in endometriosis patients, endometrial stem/progenitor cells were inappropriately shed during menstruation and reached the peritoneal to establish endometriotic implants.[43–45] Gargett et al also testified that adult human endometrium contained rare epithelial progenitors and mesenchymal stem cells (MSCs), which are likely responsible for its immense regenerative capacity and development of endometriosis.[46] In the study conducted by Chan et al, some cells from ovarian endometrioma exhibited somatic stem cell properties. Derived from ovarian endometriotic cysts, purified epithelial and stromal cells established colony-forming units, and presented with self-renewal and multi-potent capacities.[47] Meanwhile, the migration, proliferation, and angiogenic ability were more obvious in ectopic endometrial MSCs from patients with endometriosis, when compared to eutopic MSCs from the same patient or control MSCs from women without endometriosis.[48,49] Kao et al also observed ectopic MSCs with increased angiogenesis and invasion into the surrounding tissue in a scaffold transplantation mouse model.[48] Moggio et al demonstrated that sorafenib might decrease the higher migratory, proliferative, and angiogenic phenotype of ectopic MSCs, showing that MSCs are helpful for both illuminating the pathogenesis, and exploring novel non-hormonal therapy.[49]
Endometrial organoids
Organoids were stem-cell-derived structures generated in vitro, which display the 3D architecture and physiology of original organs.[50] These offer extraordinary opportunities for modeling and exploring the normal development and disease processes, and gave rise to novel approaches to drug screening and toxicology testing.[50] In recent years, organoid culture has again gained the attention of researchers and received further development. As a genetically stable and self-organizing 3D culture system, organoids contain both progenitor/stem and differentiated cells that resemble the original tissue. Human organoids have been derived from tissue-resident adult epithelial stem cells from the gut, liver, endometrium, as well as other organs.[51–53] Turco et al and Boretto et al were the first scholars to report the study of EOs, including mouse and human EOs.[53,54] Based on these, Fitzgerald et al and Boretto et al further investigated the characteristics of human EOs, including the different cell types of EOs and ectopic and eutopic EOs of endometriosis patients.[55,56] These studies explored the components of various EO culture mediums, which was usually referred to as the R-spondin-based culture method. This elucidated the characteristics of specific types of EOs at the phenotypic and genetic levels, and revealed that EOs are positive in cytokeratin, E-cadherin, steroid receptors, mucus, and intact epithelial polarity. Furthermore, this tested the responsiveness to hormones, which influenced the expression of ER and PR, gene profiles, and cell types.[53–56] Importantly, they verified that EOs could maintain the important phenotypic and genetic characteristics after long-term expansion. As an ideal model, EOs are expected to play an important role in pathophysiology research and the drug screening of endometriosis.
In Vivo Models
Compared with in vitro models, in vivo models have extraordinary advantages in exploring etiology, novel therapies, and the influence of endometriosis on patients, such as fertility or pain, which require behavior analysis. The most common animal models included non-human primates (NHPs) and rodent animals. Each category has its own merits and limitations [Table 3].
Table 3.
The merits and weaknesses of animal models.

Non-human primates
Spontaneous endometriosis only occurs in humans and NHPs, such as rhesus monkeys and baboons, which have nature menstrual cycles.[57] Endometriosis in NHPs resemble the human condition, in terms of phylogenetics, reproductive anatomy and physiology, laparoscopic and microscopic aspects.[58] Furthermore, NHPs are only species with spontaneous or induced endometriosis similar to the disease in women.[59] In the wild, endometriosis infrequently and slowly develops, which result in the establishment of an induced model via the injection of autologous menstrual effluent into the pelvic cavities of baboons.[60] In general, the presence or absence of endometriosis was checked by laparoscopy. After endometrium was injected into the abdominal cavity, laparoscopic procedures were performed at different time points to observe the endometriotic lesions and development of the disease.[61,62] The reasons why baboons were the most frequent models include noninvasive cycle monitoring based on perineal changes, continuous breeding, suitable size and strength, spontaneous peritoneal fluid, cross-reactivity between baboons and humans, vaginal transcervical uterine access, spontaneous retrograde menstruation, and human-like minimal to severe endometriosis.[59] In a classic design, D’Hooghe et al found that compared with retroperitoneal injection, intra-peritoneal implantation using menstrual endometrium, rather than luteal endometrium, could more successfully induce endometriosis.[62] Frequently, baboons have been used to explore the possible pathophysiology and potential therapies.[63,64] There is also a saying that new drugs that exhibit effective potential in the rodent model needs to be further tested, in terms of the general and reproductive side effect in NHPs.[59]
Rodent animal models
Although there are a lot of advantages in the NHP model, the limitation of price and ethical considerations make its frequent use difficult. On the contrary, rodent models are relatively more general with respect to economics, and easy to maneuver and perform genetic manipulation, such as the knock-out mice and transgenic mice.
The rat autologous model was developed by Vernon and Wilson in 1985,[65] and subsequently modified by Berkley et al,[66] who sutured small pieces of uterus not only to the mesenteric cascade, but also onto the abdomen and ovary, to further resemble the distribution of lesions in women. In addition, Prodromidou et al attempted to establish the DIE model in rats by suturing the resectied uterine horn to the rectum of rats, and confirmed this macroscopically and microscopically.[67] Except for verifying the reduced fertility,[65] the rat model was also commonly used to explore the association between endometriosis and increase in pelvic nociception, such as the vaginal hyperalgesia and muscle hyperalgesia induced by a ureteral calculosis amid behavior analysis, and this might be partly explained via the “viscero-visceral referred hyperalgesia” and central sensitization.[66,68] At the same time, some studies explored relevant therapies for “viscero-visceral hyperalgesia” and central sensitization using rat models, such as ketoprofen.[68]
Uterine fragments have also once been grafted onto the sciatic nerve to imitate neuropathic pain in endometriosis.[69] In addition to exploring the pain mechanism, a rat model was used to study novel therapies and relevant pathophysiology. For instance, cisplatin and letrozole have been tested for the treatment of endometriosis on a rat model.[70] Furthermore, the rat model was used to analyze the gene expression profiles of ectopic tissues deposited in rats, and explore the association between lesions and inflammatory response, angiogenesis, extracellular environment, and so on.[71–73] However, the establishment of rat models were mainly surgically induced by suturing fragments of uterine tissue to the peritoneum and omentum from the same or syngeneic donor.[74] The injection method did not work in rats, because the fragments failed to attach and invade the peritoneal cavity.[65] In contrast, due to the involvement of knockout and transgenic mouse, the mouse model was more various. Apart from the autologous model, there was also the patient-derived xenograft (PDX), which indicated the humanized mouse model of endometriosis by grafting intact human tissue or human endometriosis cell lines.[75] In addition, even in the syngeneic mouse models, the experimental methods were different, ranging from the suturing tissue to the peritoneal lining, and the injection of whole uterine fragments to the injection of “menstrual” material, which was described by Greaves et al in detail.[75] Among these models, the use of steroid-induced menstruation as the source of syngeneic mouse menstrual endometrium and the introduction of this into the peritoneum of immunocompetent mice[76,77] further simulated the human disease process. These different mice models were complementary to each other. For example, although human tissues could be manipulated before xenografting in heterologous models, but these cannot be used to study the immune system due to immunodeficiency. In contrast, the immunocompetent mouse model could be used to study the effect of immune-modulating drugs and anti-inflammatory agents.[75] The mouse model has also been involved in genetic manipulation, which was applied to certain target genes to investigate alterations in ERβ activity during endometriosis progression.[78]
Conclusions and Perspective
As a debilitating, chronic and recurrent disease, endometriosis affects around 6% to 10% of women in their reproductive age, and this substantially affects the quality of life of women, and imposes costs on the society, which is similar to other chronic conditions, such as type-2 diabetes mellitus, rheumatoid arthritis and Crohn disease.[7] To better understand this enigmatic disease, the establishment of reliable endometriotic models for further research are indispensable. Traditional endometriotic models include cell lines and animal models. Primary cell lines could better represent the disease, but have a limited lifespan. For immortal cell lines, EEC12 was the most widely used, others like CRL-7566 and ESC22B have also been used in research. An ideal cell line should be able to passage and maintain the original phenotype and genotype in the long-term. However, SV40 T antigen transfected cell lines usually have karyotype abnormalities.[79] Although hTERT transfection can maintain the normal karyotype, many formed cell lines do not express or express ER and/or PR only at the RNA level, when compared to the protein level, and these could not respond to hormonal stimulation.[28,35] Indeed, endometriosis is an estrogen-dependent disease. Therefore, although these cell lines are easy to culture for a long time, there are general limitations of using such cell lines, including their genetic background, potential changes occurring during transformation and culture.[80] In addition, ethical and economic reasons limit the use of NHPs. Although rodent models are valuable in research, the research outcomes are critical in view of the differences between animals and humans. For example, IFN-α-2b, which has been shown to be efficient in the treatment of rodent endometriosis, makes the endometriosis more severe in patients.[81,82]
Organoids partly solved above mentioned problems. In oncology research, researchers have found that tumor cell lines cannot retain certain important mutations, and the barcode complexity of cell lines was also progressively lost.[83] Organoids could maintain the genetic stability of the original tissue, even for tumor significant genetic heterogeneity, which is better than tumor cell lines.[84,85] Turco et al and Boretto et al concluded that EOs phenotypically and genetically resemble the original characteristics, even after long-term expansion, which are important for establishing an ideal disease model.[53,54] In addition, the transcriptomic and genetic analyses of EOs could also reveal disease-associated traits, such as the gene expression differences associated with the signaling pathway, hormonal response, and the adhesion/invasion factors exhibited among the normal EOs, eutopic EOs, and ectopic EOs of endometriosis patients.[56] Therefore, EOs can also be used as reliable disease models for pathogenesis research and drug screening, which are similar to other organ-derived organoids. Similar to drug screening for cystic fibrosis[86] and colorectal cancer,[87] endometrial cancer EOs present with patient-specific drug responses.[56] In addition to being superior to cell lines, Schutte et al discovered that the response to various drugs between parallel organoids culture and PDX was generally consistent,[88] while PDX demanded more time and resources. Therefore, before conducting clinical trials, screening out sensitive drugs by combining organoids and PDX greatly improve the efficiency of drug screening, and saves time and costs.[85]
Despite the numerous merits and potential application in clinical medicine, there are still some unresolved technical issues, such as the lack of blood vessels and immune cells in most of the present organoid protocols.[50,89] The communication between epithelial cells with stromal and immune cells play an essential role in the development of endometriosis, but present EOs models cannot solve this problem, which needs to be handled for endometriotic EOs in the future.
Endometriosis is a heterogeneous condition, and the subtypes may differ from pathogenesis and require different treatments, and even require different markers for diagnosis and stratification. Zondervan et al once put forward that future research must focus on understanding the pathogenesis, identifying disease subtypes, developing non-invasive diagnostic methods, and targeting non-hormonal treatments appropriate for women who wished to conceive.[7] An exclusive endometriosis organoids biobank for pathophysiology research and drug screening are expected to solve these problems. Taken together, all kinds of models should corporate with others. As suggested by Bredenoord et al, organoids are complementary to, rather than in competition with, these classical research methodologies.[50]
Funding
This study was supported by a grant from the National Key Research and Development Program of China (No. 2017YFC1001200).
Conflicts of interest
None.
Footnotes
How to cite this article: Gu ZY, Jia SZ, Leng JH. Establishment of endometriotic models: the past and future. Chin Med J 2020;133:1703–1710. doi: 10.1097/CM9.0000000000000885
References
- 1.Giudice LC, Kao LC. Endometriosis. Lancet 2004; 364:1789–1799. doi: 10.1016/s0140-6736(04)17403-5. [DOI] [PubMed] [Google Scholar]
- 2.Farquhar C. Endometriosis. BMJ 2007; 334:249–253. doi: 10.1136/bmj.39073.736829.BE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Garai J, Molnar V, Varga T, Koppan M, Torok A, Bodis J. Endometriosis: harmful survival of an ectopic tissue. Front Biosci 2006; 11:595–619. doi: 10.2741/1821. [DOI] [PubMed] [Google Scholar]
- 4.Bulun SE. Endometriosis. N Engl J Med 2009; 360:268–279. doi: 10.1056/NEJMra0804690. [DOI] [PubMed] [Google Scholar]
- 5.Nisolle M, Donnez J. Peritoneal endometriosis, ovarian endometriosis, and adenomyotic nodules of the rectovaginal septum are three different entities. Fertil Steril 1997; 68:585–596. doi: 10.1016/s0015-0282(97)00191-x. [DOI] [PubMed] [Google Scholar]
- 6.Pearce CL, Templeman C, Rossing MA, Lee A, Near AM, Webb PM, et al. Association between endometriosis and risk of histological subtypes of ovarian cancer: a pooled analysis of case-control studies. Lancet Oncol 2012; 13:385–394. doi: 10.1016/s1470-2045(11)70404-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zondervan KT, Becker CM, Koga K, Missmer SA, Taylor RN, Vigano P. Endometriosis. Nat Rev Dis Primers 2018; 4:9.doi: 10.1038/s41572-018-0008-5. [DOI] [PubMed] [Google Scholar]
- 8.Arosh JA, Lee J, Starzinski-Powitz A, Banu SK. Selective inhibition of prostaglandin E2 receptors EP2 and EP4 modulates DNA methylation and histone modification machinery proteins in human endometriotic cells. Mol Cell Endocrinol 2015; 409:51–58. doi: 10.1016/j.mce.2015.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Colon-Diaz M, Baez-Vega P, Garcia M, Ruiz A, Monteiro JB, Fourquet J, et al. HDAC1 and HDAC2 are differentially expressed in endometriosis. Reprod Sci 2012; 19:483–492. doi: 10.1177/1933719111432870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sinreih M, Anko M, Kene NH, Kocbek V, Rizner TL. Expression of AKR1B1, AKR1C3 and other genes of prostaglandin F2alpha biosynthesis and action in ovarian endometriosis tissue and in model cell lines. Chem Biol Interact 2015; 234:320–331. doi: 10.1016/j.cbi.2014.11.009. [DOI] [PubMed] [Google Scholar]
- 11.Pollock K, Jaraczewski TJ, Carroll MJ, Lebovic DI, Kreeger PK. Endometriotic epithelial cell response to macrophage-secreted factors is dependent on extracellular matrix context. Cell Mol Bioeng 2014; 7:409–420. doi: 10.1007/s12195-014-0339-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sha L, Huang L, Luo X, Bao J, Gao L, Pan Q, et al. Long non-coding RNA LINC00261 inhibits cell growth and migration in endometriosis. J Obstet Gynaecol Res 2017; 43:1563–1569. doi: 10.1111/jog.13427. [DOI] [PubMed] [Google Scholar]
- 13.Eggers JC, Martino V, Reinbold R, Schäfer SD, Kiesel L, Starzinski-Powitz A, et al. MicroRNA miR-200b affects proliferation, invasiveness and stemness of endometriotic cells by targeting ZEB1, ZEB2 and KLF4. Reprod Biomed Online 2016; 32:434–445. doi: 10.1016/j.rbmo.2015.12.013. [DOI] [PubMed] [Google Scholar]
- 14.Kavoussi SK, Stephen SD, Lee J, Banu SK, Arosh JA, Lebovic DI. PPAR-gamma activation inhibits proliferation of endometriotic epithelial and stromal cells and is pro-apoptotic in endometriotic stromal cells in vitro. Fertil Steril 2010; 94:S40.doi: 10.1016/j.fertnstert.2010.07.155. [Google Scholar]
- 15.Beranic N, Brozic P, Brus B, Sosic I, Gobec S, Lanisnik Rizner T. Expression of human aldo-keto reductase 1C2 in cell lines of peritoneal endometriosis: potential implications in metabolism of progesterone and dydrogesterone and inhibition by progestins. J Steroid Biochem Mol Biol 2012; 130:16–25. doi: 10.1016/j.jsbmb.2011.12.011. [DOI] [PubMed] [Google Scholar]
- 16.Dong M, Yang P, Hua F. MiR-191 modulates malignant transformation of endometriosis through regulating TIMP3. Med Sci Monit 2015; 21:915–920. doi: 10.12659/msm.893872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lakshminarasimhan R, Andreu-Vieyra C, Lawrenson K, Duymich CE, Gayther SA, Liang G, et al. Down-regulation of ARID1A is sufficient to initiate neoplastic transformation along with epigenetic reprogramming in non-tumorigenic endometriotic cells. Cancer Lett 2017; 401:11–19. doi: 10.1016/j.canlet.2017.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Gaetje R, Kotzian S, Herrmann G, Baumann R, Starzinski-Powitz A. Invasiveness of endometriotic cells in vitro. Lancet 1995; 346:1463–1464. doi: 10.1016/s0140-6736(95)92474-4. [DOI] [PubMed] [Google Scholar]
- 19.Gaetje R, Kotzian S, Herrmann G, Baumann R, Starzinski-Powitz A. Nonmalignant epithelial cells, potentially invasive in human endometriosis, lack the tumor suppressor molecule E-cadherin. Am J Pathol 1997; 150:461–467. [PMC free article] [PubMed] [Google Scholar]
- 20.Starzinski-Powitz A, Gaetje R, Zeitvogel A, Kotzian S, Handrow-Metzmacher H, Herrmann G, et al. Tracing cellular and molecular mechanisms involved in endometriosis. Hum Reprod Update 1998; 4:724–729. doi: 10.1093/humupd/4.5.724. [DOI] [PubMed] [Google Scholar]
- 21.Gaetje R, Winnekendonk DW, Ahr A, Kaufmann M. Ovarian cancer antigen CA 125 influences adhesion of human and mammalian cell lines in vitro. Clin Exp Obstet Gynecol 2002; 29:34–36. [PubMed] [Google Scholar]
- 22.Zeitvogel A, Baumann R, Starzinski-Powitz A. Identification of an invasive, N-cadherin-expressing epithelial cell type in endometriosis using a new cell culture model. Am J Pathol 2001; 159:1839–1852. doi: 10.1016/S0002-9440(10)63030-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Riehl R, Johnson K, Bradley R, Grunwald GB, Cornel E, Lilienbaum A, et al. Cadherin function is required for axon outgrowth in retinal ganglion cells in vivo. Neuron 1996; 17:837–848. doi: 10.1016/s0896-6273(00)80216-0. [DOI] [PubMed] [Google Scholar]
- 24.Banu SK, Lee J, Starzinski-Powitz A, Arosh JA. Gene expression profiles and functional characterization of human immortalized endometriotic epithelial and stromal cells. Fertil Steril 2008; 90:972–987. doi: 10.1016/j.fertnstert.2007.07.1358. [DOI] [PubMed] [Google Scholar]
- 25.Nezhat F, Datta MS, Hanson V, Pejovic T, Nezhat C, Nezhat C. The relationship of endometriosis and ovarian malignancy: a review. Fertil Steril 2008; 90:1559–1570. doi: 10.1016/j.fertnstert.2008.08.007. [DOI] [PubMed] [Google Scholar]
- 26.Gurates B, Sebastian S, Yang S, Zhou J, Tamura M, Fang Z, et al. WT1 and DAX-1 inhibit aromatase P450 expression in human endometrial and endometriotic stromal cells. J Clin Endocrinol Metab 2002; 87:4369–4377. doi: 10.1210/jc.2002-020522. [DOI] [PubMed] [Google Scholar]
- 27.Kiyono T, Foster SA, Koop JI, McDougall JK, Galloway DA, Klingelhutz AJ. Both Rb/p16INK4a inactivation and telomerase activity are required to immortalize human epithelial cells. Nature 1998; 396:84–88. doi: 10.1038/23962. [DOI] [PubMed] [Google Scholar]
- 28.Bono Y, Kyo S, Takakura M, Maida Y, Mizumoto Y, Nakamura M, et al. Creation of immortalised epithelial cells from ovarian endometrioma. Br J Cancer 2012; 106:1205–1213. doi: 10.1038/bjc.2012.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mita S, Nakakuki M, Ichioka M, Shimizu Y, Hashiba M, Miyazaki H, et al. Dienogest inhibits C-C motif chemokine ligand 20 expression in human endometriotic epithelial cells. Eur J Obstet Gynecol Reprod Biol 2017; 214:65–70. doi: 10.1016/j.ejogrb.2017.04.048. [DOI] [PubMed] [Google Scholar]
- 30.Brueggmann D, Templeman C, Starzinski-Powitz A, Rao NP, Gayther SA, Lawrenson K. Novel three-dimensional in vitro models of ovarian endometriosis. J Ovarian Res 2014; 7:17.doi: 10.1186/1757-2215-7-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lawrenson K, Lee N, Torres HA, Lee JM, Brueggmann D, Rao PN, et al. Src as a novel therapeutic target for endometriosis. Gynecol Oncol 2014; 135:100–107. doi: 10.1016/j.ygyno.2014.06.016. [DOI] [PubMed] [Google Scholar]
- 32.Matsuzaki S, Darcha C. Epithelial to mesenchymal transition-like and mesenchymal to epithelial transition-like processes might be involved in the pathogenesis of pelvic endometriosis. Hum Reprod 2012; 27:712–721. doi: 10.1093/humrep/der442. [DOI] [PubMed] [Google Scholar]
- 33.Li NF, Broad S, Lu YJ, Yang JS, Watson R, Hagemann T, et al. Human ovarian surface epithelial cells immortalized with hTERT maintain functional pRb and p53 expression. Cell Prolif 2007; 40:780–794. doi: 10.1111/j.1365-2184.2007.00462.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Boccellino M, Quagliuolo L, Verde A, La Porta R, Crispi S, Piccolo MT, et al. In vitro model of stromal and epithelial immortalized endometriotic cells. J Cell Biochem 2012; 113:1292–1301. doi: 10.1002/jcb.24000. [DOI] [PubMed] [Google Scholar]
- 35.Samalecos A, Reimann K, Wittmann S, Schulte HM, Brosens JJ, Bamberger AM, et al. Characterization of a novel telomerase-immortalized human endometrial stromal cell line, St-T1b. Reprod Biol Endocrinol 2009; 7:76.doi: 10.1186/1477-7827-7-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chiappini F, Baston JI, Vaccarezza A, Singla JJ, Pontillo C, Miret N, et al. Enhanced cyclooxygenase-2 expression levels and metalloproteinase 2 and 9 activation by Hexachlorobenzene in human endometrial stromal cells. Biochem Pharmacol 2016; 109:91–104. doi: 10.1016/j.bcp.2016.03.024. [DOI] [PubMed] [Google Scholar]
- 37.Brown DM, Lee HC, Liu S, Quick CM, Fernandes LM, Simmen FA, et al. Notch-1 signaling activation and progesterone receptor expression in ectopic lesions of women with endometriosis. J Endocr Soc 2018; 2:765–778. doi: 10.1210/js.2018-00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Barbier CS, Becker KA, Troester MA, Kaufman DG. Expression of exogenous human telomerase in cultures of endometrial stromal cells does not alter their hormone responsiveness. Biol Reprod 2005; 73:106–114. doi: 10.1095/biolreprod.104.035063. [DOI] [PubMed] [Google Scholar]
- 39.Krikun G, Mor G, Alvero A, Guller S, Schatz F, Sapi E, et al. A novel immortalized human endometrial stromal cell line with normal progestational response. Endocrinology 2004; 145:2291–2296. doi: 10.1210/en.2003-1606. [DOI] [PubMed] [Google Scholar]
- 40.Kyo S, Nakamura M, Kiyono T, Maida Y, Kanaya T, Tanaka M, et al. Successful immortalization of endometrial glandular cells with normal structural and functional characteristics. Am J Pathol 2003; 163:2259–2269. doi: 10.1016/s0002-9440(10)63583-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chapdelaine P, Kang J, Boucher-Kovalik S, Caron N, Tremblay JP, Fortier MA. Decidualization and maintenance of a functional prostaglandin system in human endometrial cell lines following transformation with SV40 large T antigen. Mol Hum Reprod 2006; 12:309–319. doi: 10.1093/molehr/gal034. [DOI] [PubMed] [Google Scholar]
- 42.Halme J, Hammond MG, Hulka JF, Raj SG, Talbert LM. Retrograde menstruation in healthy women and in patients with endometriosis. Obstet Gynecol 1984; 64:151–154. [PubMed] [Google Scholar]
- 43.Gargett CE. Stem cells in gynaecology. Aust N Z J Obstet Gynaecol 2004; 44:380–386. doi: 10.1111/j.1479-828X.2004.00290.x. [DOI] [PubMed] [Google Scholar]
- 44.Starzinski-Powitz A, Zeitvogel A, Schreiner A, Baumann R. In search of pathogenic mechanisms in endometriosis: the challenge for molecular cell biology. Curr Mol Med 2001; 1:655–664. doi: 10.2174/1566524013363168. [DOI] [PubMed] [Google Scholar]
- 45.Gargett CE. Identification and characterisation of human endometrial stem/progenitor cells. Aust N Z J Obstet Gynaecol 2006; 46:250–253. doi: 10.1111/j.1479-828X.2006.00582.x. [DOI] [PubMed] [Google Scholar]
- 46.Gargett CE, Schwab KE, Zillwood RM, Nguyen HP, Wu D. Isolation and culture of epithelial progenitors and mesenchymal stem cells from human endometrium. Biol Reprod 2009; 80:1136–1145. doi: 10.1095/biolreprod.108.075226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chan RW, Ng EH, Yeung WS. Identification of cells with colony-forming activity, self-renewal capacity, and multipotency in ovarian endometriosis. Am J Pathol 2011; 178:2832–2844. doi: 10.1016/j.ajpath.2011.02.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kao AP, Wang KH, Chang CC, Lee JN, Long CY, Chen HS, et al. Comparative study of human eutopic and ectopic endometrial mesenchymal stem cells and the development of an in vivo endometriotic invasion model. Fertil Steril 2011; 95:1308–1315.e1301. doi: 10.1016/j.fertnstert.2010.09.064. [DOI] [PubMed] [Google Scholar]
- 49.Moggio A, Pittatore G, Cassoni P, Marchino GL, Revelli A, Bussolati B. Sorafenib inhibits growth, migration, and angiogenic potential of ectopic endometrial mesenchymal stem cells derived from patients with endometriosis. Fertil Steril 2012; 98:1521–1530.e1522. doi: 10.1016/j.fertnstert.2012.08.003. [DOI] [PubMed] [Google Scholar]
- 50.Bredenoord AL, Clevers H, Knoblich JA. Human tissues in a dish: the research and ethical implications of organoid technology. Science 2017; 355: doi: 10.1126/science.aaf9414. [DOI] [PubMed] [Google Scholar]
- 51.Huch M, Gehart H, van Boxtel R, Hamer K, Blokzijl F, Verstegen MM, et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell 2015; 160:299–312. doi: 10.1016/j.cell.2014.11.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology 2011; 141:1762–1772. doi: 10.1053/j.gastro.2011.07.050. [DOI] [PubMed] [Google Scholar]
- 53.Turco MY, Gardner L, Hughes J, Cindrova-Davies T, Gomez MJ, Farrell L, et al. Long-term, hormone-responsive organoid cultures of human endometrium in a chemically defined medium. Nat Cell Biol 2017; 19:568–577. doi: 10.1038/ncb3516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Boretto M, Cox B, Noben M, Hendriks N, Fassbender A, Roose H, et al. Development of organoids from mouse and human endometrium showing endometrial epithelium physiology and long-term expandability. Development 2017; 144:1775–1786. doi: 10.1242/dev.148478. [DOI] [PubMed] [Google Scholar]
- 55.Fitzgerald HC, Dhakal P, Behura SK, Schust DJ, Spencer TE. Self-renewing endometrial epithelial organoids of the human uterus. Proc Natl Acad Sci U S A 2019; 116:23132–23142. doi: 10.1073/pnas.1915389116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Boretto M, Maenhoudt N, Luo X, Hennes A, Boeckx B, Bui B, et al. Patient-derived organoids from endometrial disease capture clinical heterogeneity and are amenable to drug screening. Nat Cell Biol 2019; 21:1041–1051. doi: 10.1038/s41556-019-0360-z. [DOI] [PubMed] [Google Scholar]
- 57.MacKenzie WF, Casey HW. Animal model of human disease. Endometriosis. Animal model: endometriosis in rhesus monkeys. Am J Pathol 1975; 80:341–344. [PMC free article] [PubMed] [Google Scholar]
- 58.D’Hooghe TM. Clinical relevance of the baboon as a model for the study of endometriosis. Fertil Steril 1997; 68:613–625. doi: 10.1016/s0015-0282(97)00277-x. [DOI] [PubMed] [Google Scholar]
- 59.D’Hooghe TM, Kyama CM, Chai D, Fassbender A, Vodolazkaia A, Bokor A, et al. Nonhuman primate models for translational research in endometriosis. Reprod Sci 2009; 16:152–161. doi: 10.1177/1933719108322430. [DOI] [PubMed] [Google Scholar]
- 60.Lebovic DI, Mwenda JM, Chai DC, Santi A, Xu X, D’Hooghe T. Peroxisome proliferator-activated receptor-(gamma) receptor ligand partially prevents the development of endometrial explants in baboons: a prospective, randomized, placebo-controlled study. Endocrinology 2010; 151:1846–1852. doi: 10.1210/en.2009-1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.D’Hooghe TM, Bambra CS, Cornillie FJ, Isahakia M, Koninckx PR. Prevalence and laparoscopic appearance of spontaneous endometriosis in the baboon (Papio anubis, Papio cynocephalus). Biol Reprod 1991; 45:411–416. doi: 10.1095/biolreprod45.3.411. [DOI] [PubMed] [Google Scholar]
- 62.D’Hooghe TM, Bambra CS, Raeymaekers BM, De Jonge I, Lauweryns JM, Koninckx PR. Intrapelvic injection of menstrual endometrium causes endometriosis in baboons (Papio cynocephalus and Papio anubis). Am J Obstet Gynecol 1995; 173:125–134. doi: 10.1016/0002-9378(95)90180-9. [DOI] [PubMed] [Google Scholar]
- 63.Joshi NR, Su RW, Chandramouli GVR, Khoo SK, Jeong JW, Young SL, et al. Altered expression of microRNA-451 in eutopic endometrium of baboons (Papio anubis) with endometriosis. Hum Reprod 2015; 30:2881–2891. doi: 10.1093/humrep/dev229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Hussein M, Chai DC, Kyama CM, Mwenda JM, Palmer SS, Gotteland J-P, et al. c-Jun NH2-terminal kinase inhibitor bentamapimod reduces induced endometriosis in baboons: an assessor-blind placebo-controlled randomized study. Fertil Steril 2016; 105:815–824.e5. doi: 10.1016/j.fertnstert.2015.11.022. [DOI] [PubMed] [Google Scholar]
- 65.Vernon MW, Wilson EA. Studies on the surgical induction of endometriosis in the rat. Fertil Steril 1985; 44:684–694. [PubMed] [Google Scholar]
- 66.Berkley KJ, Cason A, Jacobs H, Bradshaw H, Wood E. Vaginal hyperalgesia in a rat model of endometriosis. Neurosci Lett 2001; 306:185–188. doi: 10.1016/s0304-3940(01)01906-1. [DOI] [PubMed] [Google Scholar]
- 67.Prodromidou A, Pergialiotis V, Pavlakis K, Korou LM, Frountzas M, Dimitroulis D, et al. A novel experimental model of colorectal endometriosis. J Invest Surg 2018; 31:275–281. doi: 10.1080/08941939.2017.1317374. [DOI] [PubMed] [Google Scholar]
- 68.Giamberardino MA, Berkley KJ, Affaitati G, Lerza R, Centurione L, Lapenna D, et al. Influence of endometriosis on pain behaviors and muscle hyperalgesia induced by a ureteral calculosis in female rats. Pain 2002; 95:247–257. doi: 10.1016/s0304-3959(01)00405-5. [DOI] [PubMed] [Google Scholar]
- 69.Chen S, Xie W, Strong JA, Jiang J, Zhang JM. Sciatic endometriosis induces mechanical hypersensitivity, segmental nerve damage, and robust local inflammation in rats. Eur J Pain 2016; 20:1044–1057. doi: 10.1002/ejp.827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Li Z, Liu H, He Z, Zhang G, Lang J. Effects of cisplatin and letrozole on surgically induced endometriosis and comparison of the two medications in a rat model. Eur J Pharm Sci 2016; 93:132–140. doi: 10.1016/j.ejps.2016.07.018. [DOI] [PubMed] [Google Scholar]
- 71.Flores I, Rivera E, Ruiz LA, Santiago OI, Vernon MW, Appleyard CB. Molecular profiling of experimental endometriosis identified gene expression patterns in common with human disease. Fertil Steril 2007; 87:1180–1199. doi: 10.1016/j.fertnstert.2006.07.1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Konno R, Fujiwara H, Netsu S, Odagiri K, Shimane M, Nomura H, et al. Gene expression profiling of the rat endometriosis model. Am J Reprod Immunol 2007; 58:330–343. doi: 10.1111/j.1600-0897.2007.00507.x. [DOI] [PubMed] [Google Scholar]
- 73.Umezawa M, Sakata C, Tanaka N, Kudo S, Tabata M, Takeda K, et al. Cytokine and chemokine expression in a rat endometriosis is similar to that in human endometriosis. Cytokine 2008; 43:105–109. doi: 10.1016/j.cyto.2008.04.016. [DOI] [PubMed] [Google Scholar]
- 74.Grümmer R. Translational animal models to study endometriosis-associated infertility. Semin Reprod Med 2013; 31:125–132. doi: 10.1055/s-0032-1333477. [DOI] [PubMed] [Google Scholar]
- 75.Greaves E, Critchley HOD, Horne AW, Saunders PTK. Relevant human tissue resources and laboratory models for use in endometriosis research. Acta Obstet Gynecol Scand 2017; 96:644–658. doi: 10.1111/aogs.13119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Cousins FL, Murray A, Esnal A, Gibson DA, Critchley HO, Saunders PT. Evidence from a mouse model that epithelial cell migration and mesenchymal-epithelial transition contribute to rapid restoration of uterine tissue integrity during menstruation. PLoS One 2014; 9:e86378.doi: 10.1371/journal.pone.0086378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Greaves E, Cousins FL, Murray A, Esnal-Zufiaurre A, Fassbender A, Horne AW, et al. A novel mouse model of endometriosis mimics human phenotype and reveals insights into the inflammatory contribution of shed endometrium. Am J Pathol 2014; 184:1930–1939. doi: 10.1016/j.ajpath.2014.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Han SJ, Jung SY, Wu SP, Hawkins SM, Park MJ, Kyo S, et al. Estrogen receptor beta modulates apoptosis complexes and the inflammasome to drive the pathogenesis of endometriosis. Cell 2015; 163:960–974. doi: 10.1016/j.cell.2015.10.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rinehart CA, Laundon CH, Mayben JP, Lyn-Cook BD, Kaufman DG. Conditional immortalization of human endometrial stromal cells with a temperature-sensitive simian virus 40. Carcinogenesis 1993; 14:993–999. doi: 10.1093/carcin/14.5.993. [DOI] [PubMed] [Google Scholar]
- 80.Mannelli C, Ietta F, Avanzati AM, Skarzynski D, Paulesu L. Biological tools to study the effects of environmental contaminants at the feto-maternal interface. Dose Response 2015; 13:1559325815611902.doi: 10.1177/1559325815611902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ingelmo JM, Quereda F, Acien P. Intraperitoneal and subcutaneous treatment of experimental endometriosis with recombinant human interferon-alpha-2b in a murine model. Fertil Steril 1999; 71:907–911. doi: 10.1016/s0015-0282(99)00087-4. [DOI] [PubMed] [Google Scholar]
- 82.Acien P, Quereda F, Campos A, Gomez-Torres MJ, Velasco I, Gutierrez M. Use of intraperitoneal interferon alpha-2b therapy after conservative surgery for endometriosis and postoperative medical treatment with depot gonadotropin-releasing hormone analog: a randomized clinical trial. Fertil Steril 2002; 78:705–711. doi: 10.1016/s0015-0282(02)03330-7. [DOI] [PubMed] [Google Scholar]
- 83.Porter SN, Baker LC, Mittelman D, Porteus MH. Lentiviral and targeted cellular barcoding reveals ongoing clonal dynamics of cell lines in vitro and in vivo. Genome Biol 2014; 15:R75.doi: 10.1186/gb-2014-15-5-r75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Pauli C, Hopkins BD, Prandi D, Shaw R, Fedrizzi T, Sboner A, et al. Personalized in vitro and in vivo cancer models to guide precision medicine. Cancer Discov 2017; 7:462–477. doi: 10.1158/2159-8290.CD-16-1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Weeber F, Ooft SN, Dijkstra KK, Voest EE. Tumor organoids as a pre-clinical cancer model for drug discovery. Cell Chem Biol 2017; 24:1092–1100. doi: 10.1016/j.chembiol.2017.06.012. [DOI] [PubMed] [Google Scholar]
- 86.Dekkers JF, Gogorza Gondra RA, Kruisselbrink E, Vonk AM, Janssens HM, de Winter-de Groot KM, et al. Optimal correction of distinct CFTR folding mutants in rectal cystic fibrosis organoids. Eur Respir J 2016; 48:451–458. doi: 10.1183/13993003.01192-2015. [DOI] [PubMed] [Google Scholar]
- 87.van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 2015; 161:933–945. doi: 10.1016/j.cell.2015.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Schutte M, Risch T, Abdavi-Azar N, Boehnke K, Schumacher D, Keil M, et al. Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors. Nat Commun 2017; 8:14262.doi: 10.1038/ncomms14262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 2014; 345:1247125.doi: 10.1126/science.1247125. [DOI] [PubMed] [Google Scholar]
