1
|
Hoffmann-Młodzianowska M, Maksym RB, Pucia K, Kuciak M, Mackiewicz A, Kieda C. Endometriosis development in relation to hypoxia: a murine model study. Mol Med 2024; 30:195. [PMID: 39478503 DOI: 10.1186/s10020-024-00973-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Endometriosis, due to its ambiguous symptoms, still remains one of the most difficult female diseases to treat, with an average diagnosis time of 7-9 years. The changing level of hypoxia plays an important role in a healthy endometrium during menstruation and an elevated expression of the hypoxia-inducible factor 1-alpha (HIF-1α) has been demonstrated in ectopic endometria. HIF-1α mediates the induction of proangiogenic factors and the development of angiogenesis is a critical step in the establishment and pathogenesis of endometriosis. Although the inhibition of angiogenesis has been proposed as one of the actionable therapeutic modalities, vascular normalization and re-oxygenation may become a possible new approach for therapeutic intervention. METHODS Our goal was to investigate whether a selected murine model of endometriosis would be suitable for future studies on new methods for treating endometriosis. Non-invasive, high-resolution ultrasound-monitored observation was selected as the preclinical approach to obtain imaging of the presence and volume of the endometriotic-like lesions. The EF5 (2-(2-Nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl)acetamide) compound that selectively binds to reduced proteins in hypoxic cells was used for hypoxia detection. The expression of Pten and other crucial genes linking endometriosis and hypoxia were also assessed. RESULTS Using EF5, a pentafluorinated derivative of the 2-nitroimidazole that is metabolically reduced by oxygen-inhibitable nitroreductase, we confirmed that hypoxia did develop in the selected model and was detected in uterine and ectopic endometriotic lesions. Moreover, the changes in oxygen tension also influenced the expression level of significant genes related to endometriosis, like Pten, Trp53, Hif1a, Epas1, and Vegfa. Their strong modulation evidenced here is indicative of model reliability. Using high-resolution ultrasound-based imaging, we present a non-invasive method of visualization that enables the detection and observation of lesion evolution throughout the duration of the experiment, which is fundamental for further preclinical studies and treatment evaluation. CONCLUSIONS The selected model and method of visualization appear to be suitable for the study of new treatment strategies based on hypoxia alleviation and blood flow restoration.
Collapse
Affiliation(s)
- Marta Hoffmann-Młodzianowska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, 04-141, Warsaw, Poland.
| | - Radosław B Maksym
- 1st Department of Obstetrics and Gynaecology, Centre of Postgraduate Medical Education, 01-004, Warsaw, Poland
| | - Katarzyna Pucia
- Animal Experimentation Laboratory of the Center for Biostructure Research, Medical University of Warsaw, 02-106, Warsaw, Poland
| | - Monika Kuciak
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-806, Poznan, Poland
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-806, Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866, Poznan, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, 04-141, Warsaw, Poland
- Centre for Molecular Biophysics, UPR4301 CNRS, 45071, Orléans, France
| |
Collapse
|
2
|
Tang W, Zhu X, Bian L, Zhang B. Research progress of dydrogesterone in the treatment of endometriosis. Eur J Obstet Gynecol Reprod Biol 2024; 296:120-125. [PMID: 38430648 DOI: 10.1016/j.ejogrb.2024.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Endometriosis is a common gynecological disease among women of reproductive age. It is a chronic estrogen and progestin related inflammatory disease. At present, the main treatments for endometriosis are drug therapy and surgery. In drug therapy, progesterone is listed as the first-line recommendation in multinational guidelines. Dydrogesterone, as an oral reversal progesterone, can slow down the metabolism of progesterone, inhibit angiogenesis and extracellular matrix degradation to inhibit the proliferation of the ectopic endometrium, induce the atrophy of the ectopic endometrium through the pro-apoptotic pathway, and treat endometriosis through multiple mechanisms of regulating inflammatory factors to reduce inflammation. Clinically, dydrogesterone treatment of endometriosis can relieve patients' symptoms, promote fertility, be used in combination, and is safe. This article will review the mechanism and clinical application of dydrogesterone in the treatment of endometriosis.
Collapse
Affiliation(s)
- Wenlu Tang
- Department of Gynecology, Fujian Provincial Geriatric Hospital, Fuzhou 350003, Fujian Province, China; The First Clinical College of Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Xiaohong Zhu
- The First Clinical College of Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Lihong Bian
- The First Clinical College of Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Bin Zhang
- The First Clinical College of Fujian Medical University, Fuzhou 350004, Fujian Province, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China; Department of Gynecology, National Regional Medical Centre, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
3
|
Huang E, Wang X, Chen L. Regulated Cell Death in Endometriosis. Biomolecules 2024; 14:142. [PMID: 38397379 PMCID: PMC10886833 DOI: 10.3390/biom14020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Regulated cell death (RCD) represents a distinct mode of cell demise, differing from accidental cell death (ACD), characterized by specific signaling cascades orchestrated by diverse biomolecules. The regular process of cell death plays a crucial role in upholding internal homeostasis, acting as a safeguard against biological or chemical damage. Nonetheless, specific programmed cell deaths have the potential to activate an immune-inflammatory response, potentially contributing to diseases by enlisting immune cells and releasing pro-inflammatory factors. Endometriosis, a prevalent gynecological ailment, remains incompletely understood despite substantial progress in unraveling associated signaling pathways. Its complexity is intricately tied to the dysregulation of inflammatory immune responses, with various RCD processes such as apoptosis, autophagic cell death, pyroptosis, and ferroptosis implicated in its development. Notably, limited research explores the association between endometriosis and specific RCD pathways like pyroptosis and cuproptosis. The exploration of regulated cell death in the context of endometriosis holds tremendous potential for further advancements. This article thoroughly reviews the molecular mechanisms governed by regulated cell death and their implications for endometriosis. A comprehensive understanding of the regulated cell death mechanism in endometriosis has the potential to catalyze the development of promising therapeutic strategies and chart the course for future research directions in the field.
Collapse
Affiliation(s)
| | | | - Lijuan Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (E.H.)
| |
Collapse
|
4
|
Soltani-Fard E, Asadi M, Taghvimi S, Vafadar A, Vosough P, Tajbakhsh A, Savardashtaki A. Exosomal microRNAs and long noncoding RNAs: as novel biomarkers for endometriosis. Cell Tissue Res 2023; 394:55-74. [PMID: 37480408 DOI: 10.1007/s00441-023-03802-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 06/23/2023] [Indexed: 07/24/2023]
Abstract
Endometriosis is a gynecological inflammatory disorder characterized by the development of endometrial-like cells outside the uterine cavity. This disease is associated with a wide range of clinical presentations, such as debilitating pelvic pain and infertility issues. Endometriosis diagnosis is not easily discovered by ultrasound or clinical examination. Indeed, difficulties in noninvasive endometriosis diagnosis delay the confirmation and management of the disorder, increase symptoms, and place a significant medical and financial burden on patients. So, identifying specific and sensitive biomarkers for this disease should therefore be a top goal. Exosomes are extracellular vesicles secreted by most cell types. They transport between cells' bioactive molecules such as noncoding RNAs and proteins. MicroRNAs and long noncoding RNAs which are key molecules transferred by exosomes have recently been identified to have a significant role in endometriosis by modulating different proteins and their related genes. As a result, the current review focuses on exosomal micro-and-long noncoding RNAs that are involved in endometriosis disease. Furthermore, major molecular mechanisms linking corresponding RNA molecules to endometriosis development will be briefly discussed to better clarify the potential functions of exosomal noncoding RNAs in the therapy and diagnosis of endometriosis.
Collapse
Affiliation(s)
- Elahe Soltani-Fard
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Marzieh Asadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and, Technologies, Shiraz University of, Medical Sciences, Shiraz, 71362 81407, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Taghvimi
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and, Technologies, Shiraz University of, Medical Sciences, Shiraz, 71362 81407, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Department of Medical Biotechnology, School of Advanced Medical Sciences and, Technologies, Shiraz University of, Medical Sciences, Shiraz, 71362 81407, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and, Technologies, Shiraz University of, Medical Sciences, Shiraz, 71362 81407, Iran.
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Wen Y, Fan L, Pang L, Zhao T, Li R, Zhang Y, Zhang L, Yang W. NeiyiKangfu tablets control the progression of endometriosis through inhibiting RAF/MEK/ERK signal pathway by targeting RKIP. Gynecol Endocrinol 2022; 38:1136-1146. [PMID: 36592742 DOI: 10.1080/09513590.2022.2162495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND NeiyiKangfu tablets (NYKF) are widely used clinically for the treatment of endometriosis (EMS), whose mechanism of action has been extensively studied. Researchers have found that NYKF may control the development of ectopic lesions by inhibiting angiogenesis and inflammatory cytokine secretion. Nevertheless, NYKF's mechanism of action remains unclear. METHODS In the present study, the function of NYKF in the progression of EMS and the associated underlying mechanism was investigated by in vivo and in vitro experiments. EMS model mice were treated with NYKF and the pro-inflammatory factors and apoptosis of ectopic endometrium as well as RAF/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling activation were assessed. In addition, human endometriosis-derived immortalized entopic stromal (hEM15A) cells transfected with or without RAF kinase inhibitor protein (RKIP)-small-interfering RNA (siRNA) were also treated with NYKF and the proliferation, migration, apoptosis, and RAF/MEK/ERK signaling activation were measured by Cell Counting Kit-8 (CCK-8), flow cytometry, Transwell, and western blot, respectively. RESULTS Results showed that NYKF increased the expression of RKIP, inhibited RAF/MEK/ERK signaling activation, and induced apoptosis while inhibiting proliferation and migration both in EMS mice and hEM15A cells. RKIP knockdown could inhibit the effect of NYKF treatment, leading to the activation of RAF/MEK/ERK signaling and the proliferation and migration of hEM15A cells. CONCLUSIONS In conclusion, these results suggest that NYKF treatment promotes apoptosis and inhibits proliferation and migration in EMS by inhibiting the RAF/MEK/ERK signaling pathway by targeting RKIP.
Collapse
Affiliation(s)
- Yi Wen
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lingxiu Fan
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lili Pang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tingting Zhao
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruonan Li
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Zhang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liye Zhang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Yang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
6
|
Regulation of angiogenesis by microRNAs and long non-coding RNAs in endometriosis. REPRODUCTIVE AND DEVELOPMENTAL MEDICINE 2022. [DOI: 10.1097/rd9.0000000000000024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
7
|
Gołąbek-Grenda A, Olejnik A. In vitro modeling of endometriosis and endometriotic microenvironment - Challenges and recent advances. Cell Signal 2022; 97:110375. [PMID: 35690293 DOI: 10.1016/j.cellsig.2022.110375] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 11/26/2022]
Abstract
Endometriosis is a chronic condition with high prevalence in reproductive age women, defined as the growth of endometrial tissue outside the uterine cavity, most commonly on the pelvic peritoneum. The ectopic endometrial lesions exist in a unique microenvironment created by the interaction of epithelial, stromal, endothelial, glandular, and immune cell components, dominated by inflammatory, angiogenic, and endocrine signals. Current research is directed at understanding the complex microenvironment of the lesions and its relationship with different endometriosis stages, phenotypes, and disease symptoms and at the development of novel diagnostic and therapeutic concepts that minimalize the undesirable side effects of current medical management. Recreating pathophysiological cellular and molecular mechanisms and identifying clinically relevant metrics to assess drug efficacy is a great challenge for the experimental disease models. This review summarizes the complete range of available in vitro experimental systems used in endometriotic studies, which reflect the multifactorial nature of the endometriotic lesion. The article discusses the simplistic in vitro models such as primary endometrial cells and endometriotic cell lines to heterogeneous 2D co-cultures, and recently more common, 3D systems based on self-organization and controlled assembly, both in microfluidic or bioprinting methodologies. Basic research models allow studying fundamental pathological mechanisms by which menstrual endometrium adheres, invades, and establishes lesions in ectopic sites. The advanced endometriosis experimental models address the critical challenges and unsolved problems and provide an approach to drug screening and medicine discovery by mimicking the complicated behaviors of the endometriotic lesion.
Collapse
Affiliation(s)
- Agata Gołąbek-Grenda
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland
| | - Anna Olejnik
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego St., 60-627 Poznan, Poland.
| |
Collapse
|
8
|
Abudula M, Fan X, Zhang J, Li J, Zhou X, Chen Y. Ectopic Endometrial Cell-Derived Exosomal Moesin Induces Eutopic Endometrial Cell Migration, Enhances Angiogenesis and Cytosolic Inflammation in Lesions Contributes to Endometriosis Progression. Front Cell Dev Biol 2022; 10:824075. [PMID: 35557941 PMCID: PMC9086167 DOI: 10.3389/fcell.2022.824075] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/31/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Endometriosis (EMs) is the most common gynaecological disorder with its etiology and/or pathophysiology remains enigmatic. Recent studies showed that extracellular vesicles (EVs), exosomes in particular, play a critical role in developing various clinical disorders. However, the implication of exosomes in endometriosis progression has not been well elucidated. Method: The ectopic stromal cellular exosomes (eEVs) were assessed by transwell assay, scratch tests, tube formation assay, western blot, and qRT-PCR analysis. Protein expression profiles of exosomes in endometrial tissue and vaginal discharge collected from patients with EMS and healthy donors were analysed by Mass spectrometry. siRNA interference technology was used to inhibit the expression of exosomal protein for the functional analysis in in-vivo. Finally, in-vitro experiments were performed to validate the results that we observed in EMs mouse model. Results:In vitro, we discovered that eEVs improved NSC migratory potential by upregulating MMP9 expression and activity. eEVs also aided angiogenesis and elevated the expression of inflammatory cytokines in ovarian epithelial cells, according to our findings. Moesin (MSN) levels in ESC exosomes were substantially greater than in NSC exosomes (1.22e8±5.58e6 vs. 6.605e7±4.574e6, LFQ intensity), as shown by protein mass spectrometry and bioinformatics analysis. In ectopic stromal cells, ERa receptors stimulated the RhoA/Rock-2/MSN pathway. We discovered that downregulating exosomal moesin reduced NSC migration (about 3-fold change) and MMP9 expression (about 2-fold change). On the other hand, Exomsni inhibited angiogenesis and inflammatory cytokine release. In vivo the result of immunohistochemistry and immunofluorescence demonstrated that exosomal MSN substantially modified the expression of MM9, VEGFR and p-VEGFR in polyclonal lesions. In addition, we discovered an elevation in the expression of proinflammatory factors in the surrounding tissue. Conclusion: Exosomal MSN derived from ectopic stromal cells can contribute to endometriosis progression by mediating the construction of a “migration-vascularization-inflammation” loop in the ectopic environment.
Collapse
Affiliation(s)
| | - Xiaodan Fan
- Department of Gynecology, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Jing Zhang
- Medical School, Ningbo University, Ningbo, China
| | - Jiajie Li
- Department of Gynecology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo University, Ningbo, China
| | - Xiaoming Zhou
- Department of Gynecology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo University, Ningbo, China
| | - Yichen Chen
- Department of Gynecology, Ningbo Women and Children’s Hospital, Ningbo, China
- Medical School, Ningbo University, Ningbo, China
- Department of Pharmacology, Ningbo Institute of Medical Science, Ningbo, China
- *Correspondence: Yichen Chen,
| |
Collapse
|
9
|
The ischemic time window of ectopic endometrial tissue crucially determines its ability to develop into endometriotic lesions. Sci Rep 2022; 12:5625. [PMID: 35379836 PMCID: PMC8980079 DOI: 10.1038/s41598-022-09577-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/21/2022] [Indexed: 11/08/2022] Open
Abstract
Endometriosis develop from shed endometrial fragments via retrograde menstruation. This affects the survival, proliferation and vascularization of the tissue and its final ability to form endometriotic lesions. Within this study, uterine tissue samples from donor mice were precultivated for 24 h or 72 h to simulate avascular periods. Their morphology, microvessel density, apoptotic activity and expression of angiogenesis-related proteins were analyzed in vitro. The formation of endometriotic lesions in vivo was assessed after transplantation of precultivated uterine tissue samples to the abdominal wall and dorsal skinfold chambers by means of high-resolution ultrasound, intravital fluorescence microscopy, histology and immunohistochemistry. In vitro, 72-h-precultivated uterine tissue samples exhibit extensive areas of tissue necrosis and high numbers of apoptotic cells as well as a significantly reduced cell and microvessel density. These samples failed to develop into endometriotic lesions. In contrast, the 24-h-precultivated samples showed, that their early vascularization and growth in vivo was improved when compared to controls. This indicates that avascular periods have a strong impact on the survival of ectopic endometrial tissue and the chance for the development of endometriosis.
Collapse
|
10
|
Dabi Y, Suisse S, Jornea L, Bouteiller D, Touboul C, Puchar A, Daraï E, Bendifallah S. Clues for Improving the Pathophysiology Knowledge for Endometriosis Using Plasma Micro-RNA Expression. Diagnostics (Basel) 2022; 12:175. [PMID: 35054341 PMCID: PMC8774370 DOI: 10.3390/diagnostics12010175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/06/2022] [Accepted: 01/09/2022] [Indexed: 02/07/2023] Open
Abstract
The pathophysiology of endometriosis remains poorly understood. The aim of the present study was to investigate functions and pathways associated with the various miRNAs differentially expressed in patients with endometriosis. Plasma samples of the 200 patients from the prospective "ENDO-miRNA" study were analyzed and all known human miRNAs were sequenced. For each miRNA, sensitivity, specificity, and ROC AUC values were calculated for the diagnosis of endometriosis. miRNAs with an AUC ≥ 0.6 were selected for further analysis. A comprehensive review of recent articles from the PubMed, Clinical Trials.gov, Cochrane Library, and Web of Science databases was performed to identify functions and pathways associated with the selected miRNAs. In total, 2633 miRNAs were found in the patients with endometriosis. Among the 57 miRNAs with an AUC ≥ 0.6: 20 had never been reported before; one (miR-124-3p) had previously been observed in endometriosis; and the remaining 36 had been reported in benign and malignant disorders. miR-124-3p is involved in ectopic endometrial cell proliferation and invasion and plays a role in the following pathways: mTOR, STAT3, PI3K/Akt, NF-κB, ERK, PLGF-ROS, FGF2-FGFR, MAPK, GSK3B/β-catenin. Most of the remaining 36 miRNAs are involved in carcinogenesis through cell proliferation, apoptosis, and invasion. The three main pathways involved are Wnt/β-catenin, PI3K/Akt, and NF-KB. Our results provide evidence of the relation between the miRNA profiles of patients with endometriosis and various signaling pathways implicated in its pathophysiology.
Collapse
Affiliation(s)
- Yohann Dabi
- Department of Obstetrics and Reproductive Medicine, Hôpital Tenon, Sorbonne University, 4 Rue de la Chine, 75020 Paris, France; (Y.D.); (C.T.); (A.P.); (E.D.)
- Clinical Research Group (GRC) Paris 6, Centre Expert Endométriose (C3E), Sorbonne University (GRC6 C3E SU), 4 Rue de la Chine, 75020 Paris, France
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), Sorbonne University, INSERM UMR_S_938, 75020 Paris, France
| | | | - Ludmila Jornea
- Paris Brain Institute—Institut du Cerveau—ICM, Inserm U1127, CNRS UMR 7225, AP-HP—Hôpital Pitié-Salpêtrière, Sorbonne University, 4 Rue de la Chine, 75020 Paris, France;
| | - Delphine Bouteiller
- Gentoyping and Sequencing Core Facility, iGenSeq, Institut du Cerveau et de la Moelle Épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l’Hôpital, 75013 Paris, France;
| | - Cyril Touboul
- Department of Obstetrics and Reproductive Medicine, Hôpital Tenon, Sorbonne University, 4 Rue de la Chine, 75020 Paris, France; (Y.D.); (C.T.); (A.P.); (E.D.)
- Clinical Research Group (GRC) Paris 6, Centre Expert Endométriose (C3E), Sorbonne University (GRC6 C3E SU), 4 Rue de la Chine, 75020 Paris, France
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), Sorbonne University, INSERM UMR_S_938, 75020 Paris, France
| | - Anne Puchar
- Department of Obstetrics and Reproductive Medicine, Hôpital Tenon, Sorbonne University, 4 Rue de la Chine, 75020 Paris, France; (Y.D.); (C.T.); (A.P.); (E.D.)
| | - Emile Daraï
- Department of Obstetrics and Reproductive Medicine, Hôpital Tenon, Sorbonne University, 4 Rue de la Chine, 75020 Paris, France; (Y.D.); (C.T.); (A.P.); (E.D.)
| | - Sofiane Bendifallah
- Department of Obstetrics and Reproductive Medicine, Hôpital Tenon, Sorbonne University, 4 Rue de la Chine, 75020 Paris, France; (Y.D.); (C.T.); (A.P.); (E.D.)
- Clinical Research Group (GRC) Paris 6, Centre Expert Endométriose (C3E), Sorbonne University (GRC6 C3E SU), 4 Rue de la Chine, 75020 Paris, France
| |
Collapse
|
11
|
Zhou Y, Jin Y, Wang Y, Wu R. Hypoxia activates the unfolded protein response signaling network: An adaptive mechanism for endometriosis. Front Endocrinol (Lausanne) 2022; 13:945578. [PMID: 36339404 PMCID: PMC9630844 DOI: 10.3389/fendo.2022.945578] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/05/2022] [Indexed: 11/22/2022] Open
Abstract
Endometriosis (EMS) is a chronic gynecological disease that affects women of childbearing age. However, the exact cause remains unclear. The uterus is a highly vascularized organ that continuously exposes endometrial cells to high oxygen concentrations. According to the "planting theory" of EMS pathogenesis, when endometrial cells fall from the uterine cavity and retrograde to the peritoneal cavity, they will face severe hypoxic stress. Hypoxic stress remains a key issue even if successfully implanted into the ovaries or peritoneum. In recent years, increasing evidence has confirmed that hypoxia is closely related to the occurrence and development of EMS. Hypoxia-inducible factor-1α (HIF-1α) can play an essential role in the pathological process of EMS by regulating carbohydrate metabolism, angiogenesis, and energy conversion of ectopic endometrial cells. However, HIF-1α alone is insufficient to achieve the complete program of adaptive changes required for cell survival under hypoxic stress, while the unfolded protein response (UPR) responding to endoplasmic reticulum stress plays an essential supplementary role in promoting cell survival. The formation of a complex signal regulation network by hypoxia-driven UPR may be the cytoprotective adaptation mechanism of ectopic endometrial cells in unfavorable microenvironments.
Collapse
|
12
|
Nenicu A, Yordanova K, Gu Y, Menger MD, Laschke MW. Differences in growth and vascularization of ectopic menstrual and non-menstrual endometrial tissue in mouse models of endometriosis. Hum Reprod 2021; 36:2202-2214. [PMID: 34109385 DOI: 10.1093/humrep/deab139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/29/2021] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Is there a difference in the growth and vascularization between murine endometriotic lesions originating from menstrual or non-menstrual endometrial fragments? SUMMARY ANSWER Endometriotic lesions developing from menstrual and non-menstrual tissue fragments share many similarities, but also exhibit distinct differences in growth and vascularization, particularly under exogenous estrogen stimulation. WHAT IS KNOWN ALREADY Mouse models are increasingly used in endometriosis research. For this purpose, menstrual or non-menstrual endometrial fragments serve for the induction of endometriotic lesions. So far, these two fragment types have never been directly compared under identical experimental conditions. STUDY DESIGN, SIZE, DURATION This was a prospective experimental study in a murine peritoneal and dorsal skinfold chamber model of endometriosis. Endometrial tissue fragments from menstruated (n = 15) and non-menstruated (n = 21) C57BL/6 mice were simultaneously transplanted into the peritoneal cavity or dorsal skinfold chamber of non-ovariectomized (non-ovx, n = 17), ovariectomized (ovx, n = 17) and ovariectomized, estrogen-substituted (ovx+E2, n = 17) recipient animals and analyzed throughout an observation period of 28 and 14 days, respectively. PARTICIPANTS/MATERIALS, SETTING, METHODS The engraftment, growth and vascularization of the newly developing endometriotic lesions were analyzed by means of high-resolution ultrasound imaging, intravital fluorescence microscopy, histology and immunohistochemistry. MAIN RESULTS AND THE ROLE OF CHANCE Menstrual and non-menstrual tissue fragments developed into peritoneal endometriotic lesions without differences in growth, microvessel density and cell proliferation in non-ovx mice. Lesion formation out of both fragment types was markedly suppressed in ovx mice. In case of non-menstrual tissue fragments, this effect could be reversed by estrogen supplementation. In contrast, endometriotic lesions originating from menstrual tissue fragments exhibited a significantly smaller volume in ovx+E2 mice, which may be due to a reduced hormone sensitivity. Moreover, menstrual tissue fragments showed a delayed vascularization and a reduced blood perfusion after transplantation into dorsal skinfold chambers when compared to non-menstrual tissue fragments, indicating different vascularization modes of the two fragment types. To limit the role of chance, the experiments were conducted under standardized laboratory conditions. Statistical significance was accepted for a value of P < 0.05. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Endometriotic lesions were induced by syngeneic tissue transplantation into recipient mice without the use of pathological endometriotic tissue of human nature. Therefore, the results obtained in this study may not fully relate to human patients with endometriosis. WIDER IMPLICATIONS OF THE FINDINGS The present study significantly contributes to the characterization of common murine endometriosis models. These models represent important tools for studies focusing on the basic mechanisms of endometriosis and the development of novel therapeutic strategies for the treatment of this frequent gynecological disease. The presented findings indicate that the combination of different experimental models and approaches may be the most appropriate strategy to study the pathophysiology and drug sensitivity of a complex disease such as endometriosis under preclinical conditions. STUDY FUNDING/COMPETING INTEREST(S) There was no specific funding of this study. The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- A Nenicu
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - K Yordanova
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Y Gu
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - M D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - M W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
13
|
Pellicer N, Galliano D, Herraiz S, Bagger YZ, Arce JC, Pellicer A. Use of dopamine agonists to target angiogenesis in women with endometriosis. Hum Reprod 2021; 36:850-858. [PMID: 33355352 DOI: 10.1093/humrep/deaa337] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Endometriosis requires medical management during a woman's reproductive years. Most treatments aim to create a hypoestrogenic milieu, but for patients wishing to conceive, drugs that allow normal ovarian function are needed. Targeting angiogenesis, a hallmark of the disease, using dopamine agonists (DAs) is a promising strategy for endometriosis treatment. Herein, we review experimental and clinical data that investigate this concept. In experimental models of endometriosis, DAs (bromocriptine, cabergoline, quinagolide) downregulate proangiogenic and upregulate antiangiogenic pathways in inflammatory, endothelial and endometrial cells, blocking cellular proliferation and reducing lesion size. Impaired secretion of vascular endothelial growth factor (VEGF) and inactivation of its receptor type-2 are key events. VEGF inhibition also reduces nerve fiber density in lesions. In humans, quinagolide shows similar effects on lesions, and DAs reduce pain and endometrioma size. Moreover, a 20-fold downregulation of Serpin-1, the gene that encodes for plasminogen activator inhibitor 1 (PAI-1), has been observed after DAs treatment. Pentoxifylline, a PAI-1, increases pregnancy rates in women with endometriosis. Thus, the data support the use of DAs in the medical management of endometriosis to reduce lesion size and pain while maintaining ovulation. A combined approach of DAs and pentoxifylline is perhaps a smart way of targeting the disease from a completely different angle than current medical treatments.
Collapse
Affiliation(s)
- Nuria Pellicer
- Women's Health Area, La Fe University Hospital, 46026 Valencia, Spain.,IVI Foundation, 46026 Valencia, Spain.,Reproductive Medicine Research Group; Biomedical Research Institute la Fe, 46026 Valencia, Spain
| | - Daniela Galliano
- IVI Foundation, 46026 Valencia, Spain.,IVI-RMA Rome, 00197 Rome, Italy
| | - Sonia Herraiz
- IVI Foundation, 46026 Valencia, Spain.,Reproductive Medicine Research Group; Biomedical Research Institute la Fe, 46026 Valencia, Spain
| | - Yu Z Bagger
- Ferring Pharmaceuticals, 2300 Copenhagen, Denmark
| | | | - Antonio Pellicer
- IVI Foundation, 46026 Valencia, Spain.,Reproductive Medicine Research Group; Biomedical Research Institute la Fe, 46026 Valencia, Spain.,IVI-RMA Rome, 00197 Rome, Italy
| |
Collapse
|
14
|
The Effect of Novel Medical Nonhormonal Treatments on the Angiogenesis of Endometriotic Lesions. Obstet Gynecol Surv 2021; 76:281-291. [PMID: 34032860 DOI: 10.1097/ogx.0000000000000888] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Importance Irrespective of the precise mechanisms leading to endometriosis, angiogenesis is essential for the establishment and long-term proliferation of the disease. As current surgical and medical management options for women with endometriosis have substantial drawbacks and limitations, novel agents are needed and molecules targeting the angiogenic cascade could serve as potential candidates. Objective Our aim was to review current data about the role of angiogenesis in the pathophysiology of endometriosis and summarize the novel antiangiogenic agents that could be potentially used in clinical management of patients with endometriosis. Evidence Acquisition Original research and review articles were retrieved through a computerized literature search. Results Loss of balance between angiogenic activators and suppressors triggers the nonphysiological angiogenesis observed in endometriotic lesions. Several proangiogenic mediators have been identified and most of them have demonstrated increased concentrations in the peritoneal fluid and/or serum of women with endometriosis. Among the antiangiogenic molecules, anti-vascular endothelial growth factor agents, dopamine agonists, romidepsin, and statins have shown the most promising results so far. Conclusions and Relevance Given the limitations of current treatments of endometriosis, there is a need for novel, more efficient agents. Antiangiogenic molecules could be used potentially in clinical management of women with endometriosis; however, their safety and efficiency should be carefully assessed prior to that. Further large prospective trials in humans are needed before any treatment is introduced into daily clinical practice.
Collapse
|
15
|
Zhao R, Feng D, Zhuang G, Liu Y, Chi S, Zhang J, Zhou X, Zhang W, Wang H. Protein kinase CK2 participates in estrogen-mediated endothelial progenitor cell homing to endometriotic lesions through stromal cells in a stromal cell-derived factor-1- CXCR4-dependent manner. Fertil Steril 2021; 113:1067-1079.e5. [PMID: 32386617 DOI: 10.1016/j.fertnstert.2019.12.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To explore the possible mechanism of protein kinase CK2, which participates in estrogen recruitment of endothelial progenitor cells (EPCs), and its role in the angiogenesis of endometriosis lesions. DESIGN Laboratory study. SETTING University. ANIMAL(S) BALB/c mice. INTERVENTION(S) Exposure of human endometrial stromal cells (HESCs) to estrogen and CK2 inhibitor CX-4945 and endometrial stromal cells transfected with the protein kinase CK2 vector (HESC-CK2). Endometriosis models were induced by allogeneic mice transplantation of the endometrium into dorsal skinfold chambers. The mice received an IP injection of 50 mg/kg emodin per day or were treated with 100 μg/kg estrogen by SC injection once a week. MAIN OUTCOME MEASURE(S) The concentration of cytokines in cells was measured with ELISA. The migration of EPCs was examined using the scratch assay method and Transwell, a capillary tube-formation assay to determine EPC tube-forming capacity, and protein and mRNA expression with Western blot and polymerase chain reaction analyses, respectively. RESULT(S) Protein kinase CK2 participates in estrogen-mediated EPC homing to endometriotic lesions through stromal cells in a stromal cell-derived factor-1 (SDF-1)-CXCR4-dependent manner. Conditioned medium from endometrial stromal cells that were stably transfected with the protein kinase CK2 vector (HESC-CK2) or pretreated with estrogen significantly enhanced the migration and recruitment of EPCs. In contrast, conditioned medium from HESCs that were treated with CX-4945, a selective inhibitor of CK2, inhibited the mobility and viability of EPCs. Furthermore, CK2 overexpression significantly upregulated SDF-1 expression and secretion in endometrial stromal cells by activating the AKT/mTOR pathway. Moreover, treatment with the SDF-1 receptor CXCR4-specific inhibitor AMD3100 completely reversed the CK2-enhanced migration of EPCs. CONCLUSION(S) This study demonstrates that CK2 participates in estrogen-mediated EPC homing to endometriotic lesions through stromal cells in an SDF-1-CXCR4-dependent manner and may be a therapeutic target.
Collapse
Affiliation(s)
- Rong Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huang Zhong University of Science and Technology, Wuhan, People's Republic of China
| | - Dilu Feng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huang Zhong University of Science and Technology, Wuhan, People's Republic of China
| | - Guobin Zhuang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huang Zhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yan Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huang Zhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shuqi Chi
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Jun Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huang Zhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xing Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huang Zhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huang Zhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huang Zhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
16
|
Ilhan M, Ali Z, Khan IA, Taştan H, Küpeli Akkol E. The regression of endometriosis with glycosylated flavonoids isolated from Melilotus officinalis (L.) Pall. in an endometriosis rat model. Taiwan J Obstet Gynecol 2020; 59:211-219. [PMID: 32127140 DOI: 10.1016/j.tjog.2020.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Melilotus officinalis (L.) Pall. is commonly used for treating bronchitis, painful menstruation, hemorrhoids, kidney stones, ulcers of the eyes, earache, and hardening and swelling of uterus. The European Medicines Agency reported the use of M. officinalis orally against stomach ache, gastric ulcer, and disorders of the liver and uterus in folk medicine. The present study aimed to appraise the activity of M. (L.) Pall. aerial parts in endometriosis rat model. MATERIALS AND METHODS The endometriosis rat model was used to evaluate the potential activity of M. officinalis aerial parts based on its folkloric usage. The aerial parts of M. officinalis were extracted with n-hexane, ethyl acetate (EtOAc), and methanol (MeOH), respectively. The adhesion scores, endometrial foci areas, and cytokine levels were measured in all treated groups. After the biological activity studies, phytochemical studies were performed on the active extract and the fractions obtained from the active extract. RESULTS The MeOH extract significantly decreased the endometrial foci areas and cytokine levels in rats with endometriosis. Fractionation was performed on the MeOH extract to achieve bioactive molecules. Following the fractionation, the fractions obtained from the MeOH extract were tested. Fraction C showed the highest activity in the rat endometriosis model. Phytochemical investigation of the active fraction (Fraction C) resulted in isolation and elucidation of some quercetin and kaempferol glucoside derivatives. CONCLUSION Fraction C obtained from the MeOH extract of M. officinalis showed the highest activity, yielding four glycosylated flavonoids.
Collapse
Affiliation(s)
- Mert Ilhan
- Department of Pharmacognosy, Faculty of Pharmacy, Van Yüzüncü Yıl University, Tuşba, 65080, Van, Turkey; Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler, 06330, Ankara, Turkey; National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Zulfiqar Ali
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Ikhlas A Khan
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Hakkı Taştan
- Department of Biology, Faculty of Science, Gazi University, Etiler, 06330, Ankara, Turkey
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler, 06330, Ankara, Turkey.
| |
Collapse
|
17
|
Meresman GF, Götte M, Laschke MW. Plants as source of new therapies for endometriosis: a review of preclinical and clinical studies. Hum Reprod Update 2020; 27:367-392. [PMID: 33124671 DOI: 10.1093/humupd/dmaa039] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Given the disadvantages and limitations of current endometriosis therapy, there is a progressive increase in studies focusing on plant-derived agents as a natural treatment option with the intention of achieving high efficiency, avoiding adverse effects and preserving the chance for successful pregnancy. The heterogeneity of these studies in terms of evaluated agents, applied approaches and outcomes illustrates the need for an up-to-date summary and critical view on this rapidly growing field in endometriosis research. OBJECTIVE AND RATIONALE This review provides a comprehensive overview of plant-derived agents and natural treatment strategies that are under preclinical or clinical investigation and critically evaluates their potential for future endometriosis therapy. SEARCH METHODS An English language PubMed literature search was performed using variations of the terms 'endometriosis', 'natural therapy', 'herb/herbal', 'plant', 'flavonoid', 'polyphenol', 'phytochemical', 'bioactive', 'Kampo' and 'Chinese medicine'. It included both animal and human studies. Moreover, the Clinicaltrials.gov database was searched with the term 'endometriosis' for clinical trials on plant-derived agents. No restriction was set for the publication date. OUTCOMES Natural therapies can be assigned to three categories: (i) herbal extracts, (ii) specific plant-derived bioactive compounds and (iii) Chinese herbal medicine (CHM). Agents of the first category have been shown to exert anti-proliferative, anti-inflammatory, anti-angiogenic and anti-oxidant effects on endometrial cells and endometriotic lesions. However, the existing evidence supporting their use in endometriosis therapy is quite limited. The most studied specific plant-derived bioactive compounds are resveratrol, epigallocatechin-3-gallate, curcumin, puerarin, ginsenosides, xanthohumol, 4-hydroxybenzyl alcohol, quercetin, apigenin, carnosic acid, rosmarinic acid, wogonin, baicalein, parthenolide, andrographolide and cannabinoids, with solid evidence about their inhibitory activity in experimental endometriosis models. Their mechanisms of action include pleiotropic effects on known signalling effectors: oestrogen receptor-α, cyclooxygenase-2, interleukin-1 and -6, tumour necrosis factor-α, intercellular adhesion molecule-1, vascular endothelial growth factor, nuclear factor-kappa B, matrix metalloproteinases as well as reactive oxygen species (ROS) and apoptosis-related proteins. Numerous studies suggest that treatment with CHM is a good choice for endometriosis management. Even under clinical conditions, this approach has already been shown to decrease the size of endometriotic lesions, alleviate chronic pelvic pain and reduce postoperative recurrence rates. WIDER IMPLICATIONS The necessity to manage endometriosis as a chronic disease highlights the importance of identifying novel and affordable long-term safety therapeutics. For this purpose, natural plant-derived agents represent promising candidates. Many of these agents exhibit a pleiotropic action profile, which simultaneously inhibits fundamental processes in the pathogenesis of endometriosis, such as proliferation, inflammation, ROS formation and angiogenesis. Hence, their inclusion into multimodal treatment concepts may essentially contribute to increase the therapeutic efficiency and reduce the side effects of future endometriosis therapy.
Collapse
Affiliation(s)
- Gabriela F Meresman
- Institute of Biology and Experimental Medicine (IBYME-CONICET), C1428ADN Buenos Aires, Argentina
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
18
|
Malvezzi H, Marengo EB, Podgaec S, Piccinato CDA. Endometriosis: current challenges in modeling a multifactorial disease of unknown etiology. J Transl Med 2020; 18:311. [PMID: 32787880 PMCID: PMC7425005 DOI: 10.1186/s12967-020-02471-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Endometriosis is a chronic inflammatory hormone-dependent condition associated with pelvic pain and infertility, characterized by the growth of ectopic endometrium outside the uterus. Given its still unknown etiology, treatments usually aim at diminishing pain and/or achieving pregnancy. Despite some progress in defining mode-of-action for drug development, the lack of reliable animal models indicates that novel approaches are required. The difficulties inherent to modeling endometriosis are related to its multifactorial nature, a condition that hinders the recreation of its pathology and the identification of clinically relevant metrics to assess drug efficacy. In this review, we report and comment endometriosis models and how they have led to new therapies. We envision a roadmap for endometriosis research, integrating Artificial Intelligence, three-dimensional cultures and organ-on-chip models as ways to achieve better understanding of physiopathological features and better tailored effective treatments.
Collapse
Affiliation(s)
- Helena Malvezzi
- Hospital Israelita Albert Einstein, São Paulo, SP 05652-900 Brazil
| | - Eliana Blini Marengo
- Instituto Butanta- EstabilidadeBiotech Quality Control, São Paulo, SP 05503-900 Brazil
| | - Sérgio Podgaec
- Hospital Israelita Albert Einstein, São Paulo, SP 05652-900 Brazil
| | | |
Collapse
|
19
|
Rudzitis-Auth J, Fuß SA, Becker V, Menger MD, Laschke MW. Inhibition of erythropoietin-producing hepatoma receptor B4 (EphB4) signalling suppresses the vascularisation and growth of endometriotic lesions. Br J Pharmacol 2020; 177:3225-3239. [PMID: 32144768 DOI: 10.1111/bph.15044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE The development of endometriotic lesions is crucially dependent on the formation of new blood vessels. In the present study, we analysed whether this process is regulated by erythropoietin-producing hepatoma receptor B4 (EphB4) signalling. EXPERIMENTAL APPROACH We first assessed the anti-angiogenic action of the EphB4 inhibitor NVP-BHG712 in different in vitro angiogenesis assays. Then, endometriotic lesions were surgically induced in the dorsal skinfold chamber and peritoneal cavity of NVP-BHG712- or vehicle-treated BALB/c mice. This allowed to study the effect of EphB4 inhibition on their vascularisation and growth by means of intravital fluorescence microscopy, high-resolution ultrasound imaging, histology and immunohistochemistry. KEY RESULTS Non-cytotoxic doses of NVP-BHG712 suppressed the migration, tube formation and sprouting activity of both human dermal microvascular endothelial cells (HDMEC) and mouse aortic rings. Accordingly, we also detected a lower blood vessel density in NVP-BHG712-treated endometriotic lesions. This was associated with a reduced lesion growth due to a significantly lower number of proliferating stromal cells when compared to vehicle-treated controls. CONCLUSIONS AND IMPLICATIONS Inhibition of EphB4 signalling suppresses the vascularisation and growth of endometriotic lesions. Hence, EphB4 represents a promising pharmacological target for the treatment of endometriosis.
Collapse
Affiliation(s)
| | - Sophia A Fuß
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Vivien Becker
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
20
|
Abstract
Endometriosis is one of the most common benign gynecological diseases in women of reproductive age worldwide. In past decades, a number of in-vitro models have been used to investigate the pathology and therapeutic methods for the treatment of endometriosis. The current review summarized the majority of currently available in-vitro models, which utilize a variety of cell or tissues types, including endometriotic cell lines, primary endometrial stromal cells, endometrial stem cells, endometrial explants, peritoneal explants and immune cells. These cells or tissues are cultured individually, co-cultured in 2D or 3D systems with various matrices or cultured in chicken chorioallantotic membranes and amniotic membranes culture systems. These models are able to represent one or more aspects of the process of endometriosis. These models are helpful and can be used to investigate the development of endometriosis and the underlying mechanisms of this disorder in detail, and help investigators select appropriate models for their experiments. Recently, the new concept of endometriosis as a fibrotic condition will lead research to investigate the differentiation of myofibroblasts and the development of fibrosis in endometriotic lesions, which will increase the development of novel models that can be used to investigate endometriotic fibrosis.
Collapse
Affiliation(s)
- Hongjie Fan
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P.R. China
| |
Collapse
|
21
|
Positron emission tomography imaging of vascular endothelial growth factor with 64Cu-labeled bevacizumab for non-invasive diagnosis of endometriosis. JOURNAL OF ENDOMETRIOSIS AND PELVIC PAIN DISORDERS 2019. [DOI: 10.1177/2284026519891576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Introduction: Non-invasive diagnosis of endometriosis remains challenging. A promising approach for diagnosing endometriosis is the molecular imaging of vascular endothelial growth factor because angiogenesis plays a role in the establishment of endometriosis. This study aimed to evaluate the potential of copper-64-labeled bevacizumab, an anti–vascular endothelial growth factor antibody, for endometriosis imaging. Methods: Mouse endometriosis model was prepared by autologous transplantation. The vascular endothelial growth factor expression was evaluated by immunohistochemical staining. Biodistribution study and positron emission tomography imaging were performed at 1, 24, and 48 h after the injection of radiolabeled bevacizumab. Results: The immunohistochemical staining revealed that vascular endothelial growth factor is expressed around the stroma and glandular epithelial cells in the endometriosis lesion. The biodistribution study showed a high uptake of indium-111 bevacizumab in the endometriosis lesion. Positron emission tomography imaging with copper-64-labeled bevacizumab clearly visualized the endometriosis lesions at 24 and 48 h after injection. Conclusion: These results indicate the potential usefulness of copper-64-labeled bevacizumab for endometriosis imaging.
Collapse
|
22
|
Sekiguchi K, Ito Y, Hattori K, Inoue T, Hosono K, Honda M, Numao A, Amano H, Shibuya M, Unno N, Majima M. VEGF Receptor 1-Expressing Macrophages Recruited from Bone Marrow Enhances Angiogenesis in Endometrial Tissues. Sci Rep 2019; 9:7037. [PMID: 31065021 PMCID: PMC6504918 DOI: 10.1038/s41598-019-43185-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/08/2019] [Indexed: 01/08/2023] Open
Abstract
Angiogenesis is critical in maintenance of endometrial tissues. Here, we examined the role of VEGF receptor 1 (VEGFR1) signaling in angiogenesis and tissue growth in an endometriosis model. Endometrial fragments were implanted into the peritoneal wall of mice, and endometrial tissue growth and microvessel density (MVD) were determined. Endometrial fragments from wild-type (WT) mice grew slowly with increased angiogenesis determined by CD31+ MVD, peaking on Day 14. When tissues from WT mice were transplanted into VEGFR1 tyrosine kinase-knockout mice, implant growth and angiogenesis were suppressed on Day 14 compared with growth of WT implants in a WT host. The blood vessels in the implants were not derived from the host peritoneum. Immunostaining for VEGFR1 suggested that high numbers of VEGFR1+ cells such as macrophages were infiltrated into the endometrial tissues. When macrophages were deleted with Clophosome N, both endometrial tissue growth and angiogenesis were significantly suppressed. Bone marrow chimera experiments revealed that growth and angiogenesis in endometrial implants were promoted by host bone marrow-derived VEGFR1+/CD11b+ macrophages that accumulated in the implants, and secreted basic fibroblast growth factor (bFGF). A FGF receptor kinase inhibitor, PD173047 significantly reduced size of endometrial tissues and angiogenesis. VEGFR1 signaling in host-derived cells is crucial for growth and angiogenesis in endometrial tissue. Thus, VEGFR1 blockade is a potential treatment for endometriosis.
Collapse
Affiliation(s)
- Kazuki Sekiguchi
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.,Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan.,Department of Obstetrics and Gynecology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Yoshiya Ito
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.,Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Kyoko Hattori
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.,Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan.,Department of Obstetrics and Gynecology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Tomoyoshi Inoue
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.,Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Kanako Hosono
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.,Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Masako Honda
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.,Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan.,Department of Obstetrics and Gynecology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Akiko Numao
- Department of Obstetrics and Gynecology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Hideki Amano
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.,Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Masabumi Shibuya
- Gakubunkan Institute of Physiology and Medicine, Jobu University, Takasaki, Gunma, Japan
| | - Nobuya Unno
- Department of Obstetrics and Gynecology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Masataka Majima
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan. .,Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan.
| |
Collapse
|
23
|
Mattos RMD, Machado DE, Perini JA, Alessandra-Perini J, Meireles da Costa NDO, Wiecikowski AFDRDO, Cabral KMDS, Takiya CM, Carvalho RS, Nasciutti LE. Galectin-3 plays an important role in endometriosis development and is a target to endometriosis treatment. Mol Cell Endocrinol 2019; 486:1-10. [PMID: 30753853 DOI: 10.1016/j.mce.2019.02.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 12/14/2022]
Abstract
This study aimed to analyze galectin-3 importance in endometriotic lesions development and the effect of recombinant Gal-3 carbohydrate recognition domain (Gal3C) in experimental endometriosis treatment. Experimental endometriosis was induced in WT and Gal-3-/- mice. Initially developed lesions were macroscopically and histologically analyzed, including immunohistochemical analysis. Then, WT mice were treated with Gal3C for 15 days. Gal-3 deficiency and Gal3C treatment significantly impaired endometriosis development. A significant decrease in lesions implantation and size, VEGF and VEGFR-2 expression, vascular density and macrophage distribution were observed in Gal-3 absence or inhibition. A greater presence of iNOS positive cells was observed in knockout mice lesions, while the presence of Arginase positive cells was higher in the WT animal lesions. In addition, COX-2 and TGFb1 were reduced by Gal3C treatment. Data showed here indicate a relevant role of Gal-3 in endometriosis development and highlight a target of endometriosis treatment using Gal-3 inhibitor.
Collapse
Affiliation(s)
- Rômulo Medina de Mattos
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil; University Center IBMR, Laureate Universities, Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Daniel Escorsim Machado
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil; Research Laboratory of Pharmaceutical Sciences, West Zone State University - UEZO, Rio de Janeiro, RJ, Brazil
| | - Jamila Alessandra Perini
- Research Laboratory of Pharmaceutical Sciences, West Zone State University - UEZO, Rio de Janeiro, RJ, Brazil; Program of Post-graduation in Public Health and Environment, National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Jéssica Alessandra-Perini
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil; Research Laboratory of Pharmaceutical Sciences, West Zone State University - UEZO, Rio de Janeiro, RJ, Brazil
| | | | | | - Katia Maria Dos Santos Cabral
- National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Christina Maeda Takiya
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Renato Sampaio Carvalho
- Laboratory of Molecular Targets, Pharmaceutical Biotechnology Department, Faculty of Pharmacy, Federal University of Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Luiz Eurico Nasciutti
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
24
|
Qiu JJ, Lin XJ, Zheng TT, Tang XY, Zhang Y, Hua KQ. The Exosomal Long Noncoding RNA aHIF is Upregulated in Serum From Patients With Endometriosis and Promotes Angiogenesis in Endometriosis. Reprod Sci 2019; 26:1590-1602. [PMID: 30808247 DOI: 10.1177/1933719119831775] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The transfer of long noncoding RNAs (lncRNAs) via exosomes to modulate recipient cells represents an important mechanism for disease progression. Antisense hypoxia-inducible factor (aHIF) is a well-known angiogenesis-related lncRNA. Here, we aimed to investigate the clinical implications of aHIF and exosomal aHIF in endometriosis and the involvement of exosome-shuttled aHIF in endometriosis angiogenesis. STUDY DESIGN The distribution and expression of aHIF in ectopic, eutopic, and normal endometria was evaluated. Serum exosomal aHIF levels in patients with endometriosis were tested. The correlation between serum exosomal aHIF and aHIF expression in ectopic endometria was analyzed. Endometriotic cyst stromal cells (ECSCs)-derived exosomes were characterized. The internalization of exosomes by human umbilical vein endothelial cells (HUVECs) was observed. A series of in vitro assays were conducted to investigate the roles and mechanisms of exosomal aHIF in endometriosis angiogenesis. RESULTS Clinically, aHIF was highly expressed in ectopic endometria and serum exosomes in patients with endometriosis. Serum exosomal aHIF was significantly correlated to aHIF expression in matched ectopic endometria. In vitro, PKH67-labeled exosomes derived from aHIF high expression ECSCs were effectively internalized by recipient HUVECs. Notably, exosome-shuttled aHIF was transferred from ECSCs to HUVECs, which in turn elicited proangiogenic behavior in HUVECs by activating vascular endothelial growth factor (VEGF)-A, VEGF-D, and basic fibroblast growth factor, thereby facilitating endometriosis angiogenesis. CONCLUSION Our study illustrates a potential cell-cell communication between ECSCs and HUVECs in an ectopic environment, provides a novel mechanistic model explaining how ECSCs induce angiogenesis from the perspective of the "exosomal transfer of aHIF," and highlights the clinical value of circulating exosomal aHIF in endometriosis.
Collapse
Affiliation(s)
- Jun-Jun Qiu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Xiao-Jing Lin
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Ting-Ting Zheng
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Xiao-Yan Tang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Ying Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Ke-Qin Hua
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| |
Collapse
|
25
|
Panir K, Schjenken JE, Robertson SA, Hull ML. Non-coding RNAs in endometriosis: a narrative review. Hum Reprod Update 2019; 24:497-515. [PMID: 29697794 DOI: 10.1093/humupd/dmy014] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/05/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Endometriosis is a benign gynaecological disorder, which affects 10% of reproductive-aged women and is characterized by endometrial cells from the lining of the uterus being found outside the uterine cavity. However, the pathophysiological mechanisms causing the development of this heterogeneous disease remain enigmatic, and a lack of effective biomarkers necessitates surgical intervention for diagnosis. There is international recognition that accurate non-invasive diagnostic tests and more effective therapies are urgently needed. Non-coding RNA (ncRNA) molecules, which are important regulators of cellular function, have been implicated in many chronic conditions. In endometriosis, transcriptome profiling of tissue samples and functional in vivo and in vitro studies demonstrate that ncRNAs are key contributors to the disease process. OBJECTIVE AND RATIONALE In this review, we outline the biogenesis of various ncRNAs relevant to endometriosis and then summarize the evidence indicating their roles in regulatory pathways that govern disease establishment and progression. SEARCH METHODS Articles from 2000 to 2016 were selected for relevance, validity and quality, from results obtained in PubMed, MEDLINE and Google Scholar using the following search terms: ncRNA and reproduction; ncRNA and endometriosis; miRNA and endometriosis; lncRNA and endometriosis; siRNA and endometriosis; endometriosis; endometrial; cervical; ovary; uterus; reproductive tract. All articles were independently screened for eligibility by the authors. OUTCOMES This review integrates extensive information from all relevant published studies focusing on microRNAs, long ncRNAs and short inhibitory RNAs in endometriosis. We outline the biological function and synthesis of microRNAs, long ncRNAs and short inhibitory RNAs and provide detailed findings from human research as well as functional studies carried out both in vitro and in vivo, including animal models. Although variability in findings between individual studies exists, collectively, the extant literature justifies the conclusion that dysregulated ncRNAs are a significant element of the endometriosis condition. WIDER IMPLICATIONS There is a compelling case that microRNAs, long non-coding RNAs and short inhibitory RNAs have the potential to influence endometriosis development and persistence through modulating inflammation, proliferation, angiogenesis and tissue remodelling. Rapid advances in ncRNA biomarker discovery and therapeutics relevant to endometriosis are emerging. Unravelling the significance of ncRNAs in endometriosis will pave the way for new diagnostic tests and identify new therapeutic targets and treatment approaches that have the potential to improve clinical options for women with this disabling condition.
Collapse
Affiliation(s)
- Kavita Panir
- The Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - John E Schjenken
- The Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - M Louise Hull
- The Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Fertility SA, Adelaide, South Australia, Australia.,Department of Obstetrics and Gynaecology, Women's and Children's Hospital Adelaide, South Australia, Australia
| |
Collapse
|
26
|
Koninckx PR, Ussia A, Adamyan L, Wattiez A, Gomel V, Martin DC. Pathogenesis of endometriosis: the genetic/epigenetic theory. Fertil Steril 2018; 111:327-340. [PMID: 30527836 DOI: 10.1016/j.fertnstert.2018.10.013] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To study the pathophysiology of endometriosis. DESIGN Overview of observations on endometriosis. SETTING Not applicable. PATIENT(S) None. INTERVENTIONS(S) None. MAIN OUTCOME MEASURE(S) The hypothesis is compatible with all observations. RESULT(S) Endometriosis, endometrium-like tissue outside the uterus, has a variable macroscopic appearance and a poorly understood natural history. It is a hereditary and heterogeneous disease with many biochemical changes in the lesions, which are clonal in origin. It is associated with pain, infertility, adenomyosis, and changes in the junctional zone, placentation, immunology, plasma, peritoneal fluid, and chronic inflammation of the peritoneal cavity. The Sampson hypothesis of implanted endometrial cells following retrograde menstruation, angiogenic spread, lymphogenic spread, or the metaplasia theory cannot explain all observations if metaplasia is defined as cells with reversible changes and an abnormal behavior/morphology due to the abnormal environment. We propose a polygenetic/polyepigenetic mechanism. The set of genetic and epigenetic incidents transmitted at birth could explain the hereditary aspects, the predisposition, and the endometriosis-associated changes in the endometrium, immunology, and placentation. To develop typical, cystic ovarian or deep endometriosis lesions, a variable series of additional transmissible genetic and epigenetic incidents are required to occur in a cell which may vary from endometrial to stem cells. Subtle lesions are viewed as endometrium in a different environment until additional incidents occur. Typical cystic ovarian or deep endometriosis lesions are heterogeneous and represent three different diseases. CONCLUSION(S) The genetic epigenetic theory is compatible with all observations on endometriosis. Implications for treatment and prevention are discussed.
Collapse
Affiliation(s)
- Philippe R Koninckx
- Obstetrics-Gynecology, KU Leuven, Bierbeek, Belgium; Gruppo Italo Belga, Rome, Italy; Latiffa Hospital, Dubai, United Arab Emirates; Università Cattolica, Roma, Italy.
| | - Anastasia Ussia
- Gruppo Italo Belga, Rome, Italy; Università Cattolica, Roma, Italy
| | - Leila Adamyan
- Department of Operative Gynecology, Federal State Budget Institution V. I. Kulakov Research Centre for Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, and Department of Reproductive Medicine and Surgery, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Arnaud Wattiez
- Latiffa Hospital, Dubai, United Arab Emirates; Department of Obstetrics and Gynecology, University of Strasbourg, Strasbourg, France
| | - Victor Gomel
- Department of Obstetrics and Gynecology, University of British Columbia and Women's Hospital, Vancouver, British Columbia, Canada
| | - Dan C Martin
- School of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, and Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
27
|
Machado DE, Perini JA, de Mendonça EM, Branco JR, Rodrigues-Baptista KC, Alessandra-Perini J, Espíndola-Netto JM, Dos Santos TA, Coelho WS, Nasciutti LE, Sola-Penna M, Zancan P. Clotrimazole is effective for the regression of endometriotic implants in a Wistar rat experimental model of endometriosis. Mol Cell Endocrinol 2018; 476:17-26. [PMID: 29689297 DOI: 10.1016/j.mce.2018.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 01/31/2023]
Abstract
The present work aimed to evaluate molecular, angiogenic and inflammatory changes induced by clotrimazole (CTZ) on endometriosis lesions. For this, thirty female Wistar rats with surgically implanted autologous endometrium were treated with CTZ or vehicle (200 mg/kg) via esophageal gavage for 15 consecutive days. CTZ treatment significantly decreased the growth and the size of the implants, and histological examination indicated regression and atrophy, with no toxicity to the animals. The levels of the angiogenic markers VEGF and VEGFR-2 were significantly decreased in CTZ group. The treatment also promotes a reduction on PGE2 and TNF-α levels. All these effects involve the amelioration of ERK1/2, Akt, AMPK and PERK signaling upon CTZ treatment. In conclusion, CTZ promoted an overall amelioration of endometriosis in a rat model due to the anti-angiogenic properties of the drug. Therefore, our results support the proposal of a clinical trial using CTZ for the treatment of endometriosis.
Collapse
Affiliation(s)
- Daniel Escorsim Machado
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brazil; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jamila Alessandra Perini
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brazil; Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Osvaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Erika Menezes de Mendonça
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brazil; Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Osvaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Jessica Ristow Branco
- Laboratório de Oncobiologia Molecular (LabOMol), Departamento de Biotecnologia Farmacêutica (BioTecFar), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Karina Cristina Rodrigues-Baptista
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brazil; Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Osvaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Jessica Alessandra-Perini
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brazil; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jair Machado Espíndola-Netto
- Laboratório de Enzimologia e Controle do Metabolismo (LabECoM), Departamento de Biotecnologia Farmacêutica (BioTecFar), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thiago Alves Dos Santos
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brazil
| | - Wagner Santos Coelho
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brazil
| | - Luiz Eurico Nasciutti
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mauro Sola-Penna
- Laboratório de Enzimologia e Controle do Metabolismo (LabECoM), Departamento de Biotecnologia Farmacêutica (BioTecFar), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia Zancan
- Laboratório de Oncobiologia Molecular (LabOMol), Departamento de Biotecnologia Farmacêutica (BioTecFar), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
28
|
Ferlita AL, Battaglia R, Andronico F, Caruso S, Cianci A, Purrello M, Pietro CD. Non-Coding RNAs in Endometrial Physiopathology. Int J Mol Sci 2018; 19:ijms19072120. [PMID: 30037059 PMCID: PMC6073439 DOI: 10.3390/ijms19072120] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/12/2018] [Accepted: 07/14/2018] [Indexed: 12/18/2022] Open
Abstract
The Human Genome Project led to the discovery that about 80% of our DNA is transcribed in RNA molecules. Only 2% of the human genome is translated into proteins, the rest mostly produces molecules called non-coding RNAs, which are a heterogeneous class of RNAs involved in different steps of gene regulation. They have been classified, according to their length, into small non-coding RNAs and long non-coding RNAs, or to their function, into housekeeping non-coding RNAs and regulatory non-coding RNAs. Their involvement has been widely demonstrated in all cellular processes, as well as their dysregulation in human pathologies. In this review, we discuss the function of non-coding RNAs in endometrial physiology, analysing their involvement in embryo implantation. Moreover, we explore their role in endometrial pathologies such as endometrial cancer, endometriosis and chronic endometritis.
Collapse
Affiliation(s)
- Alessandro La Ferlita
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| | - Rosalia Battaglia
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| | - Francesca Andronico
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| | - Salvatore Caruso
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy.
| | - Antonio Cianci
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy.
| | - Michele Purrello
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| | - Cinzia Di Pietro
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
29
|
Wang XQ, Zhou WJ, Luo XZ, Tao Y, Li DJ. Synergistic effect of regulatory T cells and proinflammatory cytokines in angiogenesis in the endometriotic milieu. Hum Reprod 2018; 32:1304-1317. [PMID: 28383711 DOI: 10.1093/humrep/dex067] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/19/2017] [Indexed: 12/25/2022] Open
Abstract
STUDY QUESTION Do regulatory T cells (Tregs) contribute to angiogenesis in endometriosis? SUMMARY ANSWER High levels of CCL17 and CCL22 cause the recruitment of Tregs, upregulate the immunosuppression of Tregs and, in turn, may promote angiogenesis in endometrial cells in synergy with proinflammatory cytokines. WHAT IS ALREADY KNOWN The peritoneal fluid of patients with endometriosis has a higher percentage of Tregs than that of normal individuals; however, the regulatory role of Tregs in the disease remains unclear. STUDY DESIGN, SIZE, DURATION This study used primary human endometrial stromal cells (ESCs), monocytes (Mo), Tregs and human umbilical vein endothelial cells (HUVECs). All experiments were performed at least three times. PARTICIPANTS/MATERIALS, SETTING, METHODS The migration of Tregs was evaluated by the transwell migration assay. The activation of extracellular signal-regulated kinase (ERK)1/2, c-Jun N-terminal kinase and p38 signaling pathways was examined using the In-Cell WesternTM (LI-COR®) western blot analysis system, as well as by traditional western blot analysis. Changes in the expression of CCL22, CCL17, transforming growth factor-beta 1 (TGF-β1), Interleukin (IL)-1β, tumor necrosis factor alpha (TNF-α), IL-8 and vascular endothelial growth factor (VEGF) in cell-culture supernatant were detected by ELISA. We analyzed the Tregs by multicolor flow cytometry to directly test the expression of CCR4, CD4, CD25, Foxp3, CTLA-4, CD39 and CD73. MAIN RESULTS AND THE ROLE OF CHANCE Our results showed that ESCs-Mo co-culture produced significantly higher levels of CCL22 and CCL17 than ESCs or Mo cultured alone, and that estradiol (E2) or progesterone (P) further promoted this upregulation, demonstrating stronger chemotaxis on Tregs. The co-culture of ESCs with Mo stimulated TGF-β1 secretion by Tregs, which could be inhibited by anti-CCL22 or/and anti-CCL17 neutralizing antibodies (Abs). The expression of CCR4 by Tregs was upregulated in ESCs-Mo co-culture, especially by treatment with E2 and/or P, and this effect could be abolished by anti-CCL22 and/or anti-CCL17-neutralizing Abs. The Treg-ESCs-Mo co-culture treated with E2 (10-8 mol/l) and P (10-8 mol/l) could enhance the immunosuppression of Tregs, as proved by the elevated expression of Foxp3, CTLA-4, CD39 and CD73 on Tregs. ESCs-Mo co-culture could significantly promote the secretion of IL-1β and TNF-α. TGF-β1 from Tregs could activate p38/ERK1/2 signaling pathways in ESCs, and IL-1β and TNF-α produced by ESCs-Mo co-culture had synergistic roles with TGF-β1. TGF-β1 and the proinflammatory cytokines IL-1β or TNF-α could synergistically promote IL-8 and VEGF expression in ESCs via the p38/ERK1/2 signaling pathways. The high levels of IL-8 and VEGF in the supernatant of ESCs stimulated the angiogenesis of HUVECs. LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION This study was only performed in vitro using eutopic ESCs, instead of ectopic cells, from endometriosis patients. Therefore, it is necessary to do further experiments to determine whether Tregs promote angiogenesis in the endometriotic milieu in synergy with proinflammatory cytokines in vivo. WIDER IMPLICATIONS OF THE FINDINGS Co-targeting Tregs and proinflammatory cytokines may be an effective treatment for endometriosis. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by Ministry of Science and Technology of China 2015CB943300 to L.D.-J.; National Natural Science Foundation of China, item number 81200425 to W.X.-Q.; National Natural Science Foundation of China, item number 81471548 to L.D.-J.; and The Research Fund for the Doctoral Program of Higher Education of China to W.X.-Q. (20110071120093). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Xiao-Qiu Wang
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, IRD, Fudan University Shanghai Medical College, Shanghai, China
| | - Wen-Jie Zhou
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, IRD, Fudan University Shanghai Medical College, Shanghai, China
| | - Xue-Zhen Luo
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, IRD, Fudan University Shanghai Medical College, Shanghai, China
| | - Yu Tao
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, IRD, Fudan University Shanghai Medical College, Shanghai, China
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital and Institute of Obstetrics and Gynecology, IRD, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
30
|
Rudzitis-Auth J, Nickels RM, Menger MD, Laschke MW. Inhibition of Cyclooxygenase-2 Suppresses the Recruitment of Endothelial Progenitor Cells in the Microvasculature of Endometriotic Lesions. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:450-460. [DOI: 10.1016/j.ajpath.2017.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/04/2017] [Accepted: 10/03/2017] [Indexed: 11/16/2022]
|
31
|
Laschke MW, Menger MD. Basic mechanisms of vascularization in endometriosis and their clinical implications. Hum Reprod Update 2018; 24:207-224. [PMID: 29377994 DOI: 10.1093/humupd/dmy001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/19/2017] [Accepted: 01/01/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Vascularization is a major hallmark in the pathogenesis of endometriosis. An increasing number of studies suggests that multiple mechanisms contribute to the vascularization of endometriotic lesions, including angiogenesis, vasculogenesis and inosculation. OBJECTIVE AND RATIONALE In this review, we provide an overview of the basic mechanisms of vascularization in endometriosis and give special emphasis on their future clinical implications in the diagnosis and therapy of the disease. SEARCH METHODS Literature searches were performed in PubMed for English articles with the key words 'endometriosis', 'endometriotic lesions', 'angiogenesis', 'vascularization', 'vasculogenesis', 'endothelial progenitor cells' and 'inosculation'. The searches included both animal and human studies. No restriction was set for the publication date. OUTCOMES The engraftment of endometriotic lesions is typically associated with angiogenesis, i.e. the formation of new blood vessels from pre-existing ones. This angiogenic process underlies the complex regulation by angiogenic growth factors and hormones, which activate intracellular pathways and associated signaling molecules. In addition, circulating endothelial progenitor cells (EPCs) are mobilized from the bone marrow and recruited into endometriotic lesions, where they are incorporated into the endothelium of newly developing microvessels, referred to as vasculogenesis. Finally, preformed microvessels in shed endometrial fragments inosculate with the surrounding host microvasculature, resulting in a rapid blood supply to the ectopic tissue. These vascularization modes offer different possibilities for the establishment of novel diagnostic and therapeutic approaches. Angiogenic growth factors and EPCs may serve as biomarkers for the diagnosis and classification of endometriosis. Blood vessel formation and mature microvessels in endometriotic lesions may be targeted by means of anti-angiogenic compounds and vascular-disrupting agents. WIDER IMPLICATIONS The establishment of vascularization-based approaches in the management of endometriosis still represents a major challenge. For diagnostic purposes, reliable angiogenic and vasculogenic biomarker panels exhibiting a high sensitivity and specificity must be identified. For therapeutic purposes, novel compounds selectively targeting the vascularization of endometriotic lesions without inducing severe side effects are required. Recent progress in the field of endometriosis research indicates that these goals may be achieved in the near future.
Collapse
Affiliation(s)
- Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| |
Collapse
|
32
|
Florio P, Pinzauti S, Altomare A, Luisi S, Litta P, Petraglia F. Expression and Role of Peptides, Proteins and Growth Factors in the Pathogenesis of Endometriosis. ACTA ACUST UNITED AC 2018. [DOI: 10.1177/228402650900100203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Growing evidence is demonstrating that several peptides (corticotrophin-releasing factor, urocortins, ghrelin), proteins (leptin, adiponectin) and growth factors (vascular endothelial growth factor; epidermal growth factor family of growth factors and receptors, fibroblast growth factor, insulin like growth factor and insulin like growth factor-binding proteins, transforming growth factor-β and, activin A and related proteins) are expressed in endometriotic implants, and locally play a relevant role in affecting the biological mechanisms leading to endometriosis. They establish a complex network of interactions by which they are therefore able to stimulate angiogenesis, inflammatory cell recruitment and reaction, the growth of endometriotic tissue and its survival through the modulation of the narrow immune system. This review will evaluate the role played by several regulatory peptides, proteins and growth factors in affecting endometrial physiology and the putative mechanisms advocated to explain endometriosis (angiogenesis, cellular and humoral immunity, inflammatory response, endometrial cell proliferation, activation, motility, adhesion and invasion).
Collapse
Affiliation(s)
- Pasquale Florio
- Department of Pediatrics, Obstetrics and Reproductive Medicine, Section of Obstetrics & Gynecology, University of Siena, Siena - Italy
| | - Serena Pinzauti
- Department of Pediatrics, Obstetrics and Reproductive Medicine, Section of Obstetrics & Gynecology, University of Siena, Siena - Italy
| | - Aldo Altomare
- Department of Pediatrics, Obstetrics and Reproductive Medicine, Section of Obstetrics & Gynecology, University of Siena, Siena - Italy
| | - Stefano Luisi
- Department of Pediatrics, Obstetrics and Reproductive Medicine, Section of Obstetrics & Gynecology, University of Siena, Siena - Italy
| | - Pietro Litta
- Department of Gynecological Sciences and Human Reproduction, University of Padua, Padua - Italy
| | - Felice Petraglia
- Department of Pediatrics, Obstetrics and Reproductive Medicine, Section of Obstetrics & Gynecology, University of Siena, Siena - Italy
| |
Collapse
|
33
|
Pathogenesis of deep endometriosis. Fertil Steril 2017; 108:872-885.e1. [DOI: 10.1016/j.fertnstert.2017.08.036] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/22/2017] [Accepted: 08/28/2017] [Indexed: 12/16/2022]
|
34
|
Notch signaling controls sprouting angiogenesis of endometriotic lesions. Angiogenesis 2017; 21:37-46. [PMID: 28993956 DOI: 10.1007/s10456-017-9580-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/05/2017] [Indexed: 10/18/2022]
Abstract
Angiogenesis is essential for the engraftment and growth of endometriotic lesions. In this study, we analyzed whether this process is regulated by Notch signaling. Endometriotic lesions were induced by endometrial tissue transplantation into dorsal skinfold chambers of C57BL/6 mice, which were treated with the γ-secretase inhibitor DAPT or vehicle. Vascularization, morphology, and proliferation of the newly developing lesions were analyzed using intravital fluorescence microscopy, histology, and immunohistochemistry over 14 days. Inhibition of Notch signaling by DAPT significantly increased the number of angiogenic sprouts within the endometrial grafts during the first days after transplantation when compared to vehicle-treated controls. This was associated with an accelerated vascularization, as indicated by a higher functional microvessel density of DAPT-treated lesions on day 6. However, inhibition of Notch signaling did not affect the morphology and proliferating activity of the lesions, as previously described for tumors. Both DAPT- and vehicle-treated lesions finally consisted of cyst-like dilated glands, which were surrounded by a well-vascularized stroma and contained comparable numbers of proliferating cell nuclear antigen-positive cells. These findings demonstrate that sprouting angiogenesis in endometriotic lesions is controlled by Notch signaling. However, inhibition of Notch signaling does not have beneficial therapeutic effects on lesion development.
Collapse
|
35
|
Chen X, Jiang Y, Pan D. miR-30c may serve a role in endometriosis by targeting plasminogen activator inhibitor-1. Exp Ther Med 2017; 14:4846-4852. [PMID: 29201189 PMCID: PMC5704271 DOI: 10.3892/etm.2017.5145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/28/2016] [Indexed: 01/03/2023] Open
Abstract
The present study aimed to investigate the role of miR-30c in endometriosis (EMs) and the underlying mechanism. The expression of miR-30c and plasminogen activator inhibitor type 1 (PAI-1) mRNA in EMs tissues was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and the expression of PAI-1 protein was detected by western blot analysis. The proliferation, migration, invasion and adhesion of endometrial stromal cells (ESCs) in different groups transfected with miR-30c mimic or inhibitor were compared. It was demonstrated that miR-30c expression in ectopic and eutopic endometriosis tissues were significantly lower than in normal endometrial tissue. However, PAI-1 mRNA expression in ectopic and eutopic endometrial tissues was higher than in normal endometrial tissues. Furthermore, the expression of PAI-1 protein was higher in ectopic and eutopic endometrosis tissues than in normal tissues. RT-qPCR results indicated that miR-30c expression was significantly increased or decreased in ESCs following transfection of mimic or inhibitor of miR-30c, respectively. Overexpression of miR-30c repressed the expression of PAI-1 mRNA and protein, while inhibition of miR-30c upregulated the expression of PAI-1 in ESCs. In addition, the invasion, migration, proliferation and adhesion of ESCs was repressed following the overexpression of miR-30c, whereas they were promoted when miR-30c expression was downregulated. The results of the present study indicated that miR-30c serves an important role in the development and progression of EMs by regulating the expression of PAI-1.
Collapse
Affiliation(s)
- Xiaoli Chen
- Department of Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Yan Jiang
- Department of Gynecology and Obstetrics, Tengzhou Maternity and Child Care Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Dianling Pan
- Department of Gynecology and Obstetrics, Jinan Maternity and Child Care Hospital, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
36
|
Chang HJ, Yoo JY, Kim TH, Fazleabas AT, Young SL, Lessey BA, Jeong JW. Overexpression of Four Joint Box-1 Protein (FJX1) in Eutopic Endometrium From Women With Endometriosis. Reprod Sci 2017; 25:207-213. [PMID: 28673206 DOI: 10.1177/1933719117716780] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The four jointed box 1 (FJX1) is a regulator of angiogenesis, and the levels of FJX1 are increased in several types of cancer. Angiogenesis plays a critical role in endometrial growth as well as in several gynecologic disorders including endometriosis. However, the function of FJX1 has not been studied in endometriosis. Therefore, we examined the levels of FJX1 in eutopic endometrium from women with or without endometriosis. The levels of FJX1 protein did not change in endometrial cells during the menstrual cycle in endometrium from women without endometriosis. However, its levels were significantly higher in the secretory phase of the eutopic endometrium from women with endometriosis when compared to women without endometriosis. Hypoxia-inducible factor-1α (HIF1α) is known as a key mediator of endometriosis by regulating genes essential to estrogen production, angiogenesis, proliferation, inflammation, and extracellular invasion. It has been reported that FJX1 induces an increase in HIF1α through posttranslational stabilization. The results of our Western blot analysis reveal a significant positive correlation between FJX1 and HIF1α proteins in endometrium of women with and without endometriosis. This overexpression of FJX1 was confirmed by sequential analysis of the eutopic endometrium during endometriosis progression, using an induced model of endometriosis in the baboon. Therefore, our results suggest that high levels of FJX1 proteins may play an important role in the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Hye Jin Chang
- 1 Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA.,2 Health Promotion Center, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jung-Yoon Yoo
- 1 Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Tae Hoon Kim
- 1 Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Asgerally T Fazleabas
- 1 Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Steven L Young
- 3 Obstetrics and Gynecology, University of North Carolina, Chapel Hill, NC, USA
| | - Bruce A Lessey
- 4 Obstetrics and Gynecology, Greenville Health System, Greenville, SC, USA
| | - Jae-Wook Jeong
- 1 Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
37
|
Machado DE, Rodrigues-Baptista KC, Alessandra-Perini J, Soares de Moura R, dos Santos TA, Pereira KG, Marinho da Silva Y, Souza PJC, Nasciutti LE, Perini JA. Euterpe oleracea Extract (Açaí) Is a Promising Novel Pharmacological Therapeutic Treatment for Experimental Endometriosis. PLoS One 2016; 11:e0166059. [PMID: 27851787 PMCID: PMC5113045 DOI: 10.1371/journal.pone.0166059] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 10/21/2016] [Indexed: 12/23/2022] Open
Abstract
This study investigated the therapeutic potential of Euterpe oleracea extract (açaí) on the growth and survival of endometriotic lesions using an experimental model. Twenty female Sprague-Dawley rats were randomized into two groups after the implantation and establishment of autologous endometrium onto the peritoneum abdominal wall and treated with 200 mg/kg hydroalcoholic solution extract from açaí stone or vehicle via gastric tube for 30 consecutive days. Body weight, lesion surface areas, histological and immunohistochemistry analyses of vascular endothelial growth factor (VEGF), VEGF receptor-2 (VEGFR-2), metalloproteinase-9 (MMP-9), cyclooxygenase-2 (COX-2) and F4-80 were performed. Levels of VEGF, VEGFR-2, MMP-9 and COX-2 mRNA were measured. Flow cytometry of F4-80 was performed, and ELISA immunoassays measured prostaglandin E2 (PGE2), VEGF and nitric oxide (NO) and concentrations. Macrophage cell line J774.G8 was treated with 10, 20, and 40 μg/mL of açaí for 24, 48 and 72 h, and cell viability was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays. Açaí treatment significantly decreased the implant size, and histological examination indicated atrophy and regression. A reduction in immunostaining and mRNA expression of VEGF, MMP-9 and COX-2 was observed, and F4-80 was lower in the treated group than the control group. The treated group also exhibited lower concentrations of PGE2, VEGF and NO compared to the control group. Macrophages cells treated with 20 and 40 μg/ml of açaí reduced cell viability in about 50% after 24, 48 and 72 h. Our results suggest that açaí effectively suppressed the establishment and growth of endometriotic lesions, and this agent is a promising novel pharmacological therapeutic treatment for endometriosis.
Collapse
Affiliation(s)
- Daniel Escorsim Machado
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
| | - Karina Cristina Rodrigues-Baptista
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
- Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Osvaldo Cruz, Rio de Janeiro, RJ, Brasil
| | - Jessica Alessandra-Perini
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
- Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Osvaldo Cruz, Rio de Janeiro, RJ, Brasil
| | - Roberto Soares de Moura
- Departamento de Farmacologia e Psicobiologia, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Thiago Alves dos Santos
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
- Programa de Pós-Graduação em Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Kariny Gomes Pereira
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
| | - Yasmin Marinho da Silva
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
| | | | - Luiz Eurico Nasciutti
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Jamila Alessandra Perini
- Unidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
- Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Osvaldo Cruz, Rio de Janeiro, RJ, Brasil
- * E-mail:
| |
Collapse
|
38
|
Perelló M, González-Foruria I, Castillo P, Martínez-Florensa M, Lozano F, Balasch J, Carmona F. Oral Administration of Pentoxifylline Reduces Endometriosis-Like Lesions in a Nude Mouse Model. Reprod Sci 2016; 24:911-918. [DOI: 10.1177/1933719116673198] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Maria Perelló
- Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Iñaki González-Foruria
- Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Paola Castillo
- Biomedical Diagnostic Center, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Mario Martínez-Florensa
- Group of Immunoreceptors of the Innate and Adaptive System, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Francisco Lozano
- Group of Immunoreceptors of the Innate and Adaptive System, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Immunology Service, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
- Department of Cell Biology, Immunology and Neurosciences, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Juan Balasch
- Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
- Group of Endocrinology, Gynecology and Human Reproduction, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Francisco Carmona
- Clinical Institute of Gynecology, Obstetrics, and Neonatology, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
- Group of Endocrinology, Gynecology and Human Reproduction, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
39
|
Attar R, Attar E. Experimental Treatments of Endometriosis. WOMENS HEALTH 2016; 11:653-64. [DOI: 10.2217/whe.15.51] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Rukset Attar
- Yeditepe University Medical School, İnönü Man., Kayişdaği Cad., 26 Ağustos Yerleşimi, 34755 Ataşehir/İstanbul, Turkey
| | - Erkut Attar
- Istanbul University Istanbul Medical School, Topkapi Mh., Millet Caddesi, Çapa, 34093 Fatih/İstanbul, Turkey
| |
Collapse
|
40
|
Laschke MW, Menger MD. The gut microbiota: a puppet master in the pathogenesis of endometriosis? Am J Obstet Gynecol 2016; 215:68.e1-4. [PMID: 26901277 DOI: 10.1016/j.ajog.2016.02.036] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/01/2016] [Accepted: 02/13/2016] [Indexed: 02/08/2023]
Abstract
Endometriosis is a frequent gynecologic disease with a complex, multifactorial cause. It is characterized by the cyclic estrogen-driven proliferation and bleeding of endometriotic lesions (ie, ectopic endometrial glands and stroma) outside the uterus. These lesions induce a chronic activation of the innate immune system within the peritoneal cavity that is associated with the release of various inflammatory cytokines and angiogenic growth factors into the peritoneal fluid. This stimulates angiogenesis and the further spread of the lesions and triggers the typical pain that is symptomatic of the disease. Moreover, circulating stem and progenitor cells are recruited into the ectopic endometrial tissue and contribute to its growth and vascularization. In recent years, an increasing number of studies have indicated that the gut microbiota is not only essential for a physiologic gastrointestinal function but acts as a central regulator of a variety of inflammatory and proliferative conditions. Besides, the gut flora affects estrogen metabolism and stem-cell homeostasis. Based on these findings, we hypothesize that the gut microbiota may be involved crucially in the onset and progression of endometriosis. In the future, this novel view of the pathogenesis of endometriosis may be verified by analysis of the development of endometriotic lesions in animal models with a defined composition of the gut microbiota and by investigation of the microbiota of patients with endometriosis with modern next-generation sequencing tools. This could open the door for completely new preventive, diagnostic, and therapeutic approaches for endometriosis.
Collapse
|
41
|
Estrogen Stimulates Homing of Endothelial Progenitor Cells to Endometriotic Lesions. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2129-2142. [PMID: 27315780 DOI: 10.1016/j.ajpath.2016.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 02/29/2016] [Accepted: 04/12/2016] [Indexed: 01/19/2023]
Abstract
The incorporation of endothelial progenitor cells (EPCs) into microvessels contributes to the vascularization of endometriotic lesions. Herein, we analyzed whether this vasculogenic process is regulated by estrogen. Estrogen- and vehicle-treated human EPCs were analyzed for migration and tube formation. Endometriotic lesions were induced in irradiated FVB/N mice, which were reconstituted with bone marrow from FVB/N-TgN (Tie2/green fluorescent protein) 287 Sato mice. The animals were treated with 100 μg/kg β-estradiol 17-valerate or vehicle (control) over 7 and 28 days. Lesion growth, cyst formation, homing of green fluorescent protein(+)/Tie2(+) EPCs, vascularization, cell proliferation, and apoptosis were analyzed by high-resolution ultrasonography, caliper measurements, histology, and immunohistochemistry. Numbers of blood circulating EPCs were assessed by flow cytometry. In vitro, estrogen-treated EPCs exhibited a higher migratory and tube-forming capacity when compared with controls. In vivo, numbers of circulating EPCs were not affected by estrogen. However, estrogen significantly increased the number of EPCs incorporated into the lesions' microvasculature, resulting in an improved early vascularization. Estrogen further stimulated the growth of lesions, which exhibited massively dilated glands with a flattened layer of stroma. This was mainly because of an increased glandular secretory activity, whereas cell proliferation and apoptosis were not markedly affected. These findings indicate that vasculogenesis in endometriotic lesions is dependent on estrogen, which adds a novel hormonally regulated mechanism to the complex pathophysiology of endometriosis.
Collapse
|
42
|
Santulli P, Marcellin L, Chouzenoux S, Boulard V, Just PA, Nicco C, Chereau C, Tosti C, Chapron C, Batteux F. Role of the protein kinase BRAF in the pathogenesis of endometriosis. Expert Opin Ther Targets 2016; 20:1017-29. [PMID: 27087167 DOI: 10.1080/14728222.2016.1180367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Mitogen-activated protein kinases (MAPKs) are involved in the proliferation and survival of endometriotic lesions. Vemurafenib (PLX4032) is a novel protein kinase inhibitor that targets BRAF, a member of the MAPK pathway. The present study tested the in vitro and in vivo effects of PLX4032 on endometriotic cells. RESEARCH DESIGN AND METHODS We conducted a laboratory study in a tertiary-care university hospital from January 2013 to September 2013. We enrolled a cohort of 40 patients: 20 with histologically proven endometriosis and 20 unaffected women. A thorough surgical examination of the abdominopelvic cavity was performed on all of the study participants. Ex vivo stromal and epithelial cells were extracted from endometrial and endometriotic biopsies from both sets of patients. Proliferation, apoptosis, pERK/ERK ratio, cell cycle regulation (Cyclin D1 and CDK4) and inflammation (PTGS2) were explored with and without PLX4032 treatment. Human endometriotic lesions were implanted into 40 nude mice that were separated into two groups according to PLX4032 or vehicle treatment, which they received for four weeks, before sacrifice and histological examination. RESULTS Treating endometriotic cells with PLX4032 abrogated the phosphorylation of ERK, significantly reducing the pERK/ERK ratio in both epithelial and stromal cells from endometriotic women compared to the controls (p < 0.05). In addition, treatment with PLX4032 significantly decreased proliferation in both stromal and epithelial cells with a concomitant decrease in Cyclin D1/CDK4 complex and PTGS2 levels. Using a murine model of endometriosis, we observed that PLX4032-treated mice displayed a significant decrease in implant volume compared to the initial size; a slight, but non-significant, increase in size was observed in the vehicle-treated mice. CONCLUSION Our data suggest that MAPKs and BRAF are involved in the pathogenesis of endometriosis. PLX4032-induced inhibition of BRAF controlled endometriotic growth, both in vitro and in vivo, and could constitute a promising target for the treatment of endometriosis.
Collapse
Affiliation(s)
- Pietro Santulli
- a Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique - Hôpitaux de Paris (AP-HP), Groupe Hospitalier Universitaire (GHU) Ouest, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine , Université Paris Descartes , Paris , France.,b Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016, Equipe Pr Batteux , Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Louis Marcellin
- a Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique - Hôpitaux de Paris (AP-HP), Groupe Hospitalier Universitaire (GHU) Ouest, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine , Université Paris Descartes , Paris , France.,b Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016, Equipe Pr Batteux , Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Sandrine Chouzenoux
- b Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016, Equipe Pr Batteux , Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Veronique Boulard
- b Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016, Equipe Pr Batteux , Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | | | - Carole Nicco
- b Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016, Equipe Pr Batteux , Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Christiane Chereau
- b Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016, Equipe Pr Batteux , Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Claudia Tosti
- d Obstetrics and Gynecology, Department of Molecular and Developmental Medicine , University of Siena , Siena , Italy
| | - Charles Chapron
- a Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique - Hôpitaux de Paris (AP-HP), Groupe Hospitalier Universitaire (GHU) Ouest, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine , Université Paris Descartes , Paris , France.,b Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016, Equipe Pr Batteux , Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Frédéric Batteux
- b Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016, Equipe Pr Batteux , Université Paris Descartes, Sorbonne Paris Cité , Paris , France.,e Department of Immunology , Hôpital Cochin, AP-HP , Paris , France
| |
Collapse
|
43
|
Olivares CN, Alaniz LD, Menger MD, Barañao RI, Laschke MW, Meresman GF. Inhibition of Hyaluronic Acid Synthesis Suppresses Angiogenesis in Developing Endometriotic Lesions. PLoS One 2016; 11:e0152302. [PMID: 27018976 PMCID: PMC4809563 DOI: 10.1371/journal.pone.0152302] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/12/2016] [Indexed: 01/11/2023] Open
Abstract
Background The development and long-term survival of endometriotic lesions is crucially dependent on an adequate vascularization. Hyaluronic acid (HA) through its receptor CD44 has been described to be involved in the process of angiogenesis. Objective To study the effect of HA synthesis inhibition using non-toxic doses of 4-methylumbelliferone (4-MU) on endometriosis-related angiogenesis. Materials and Methods The cytotoxicity of different in vitro doses of 4-MU on endothelial cells was firstly tested by means of a lactate dehydrogenase assay. The anti-angiogenic action of non-cytotoxic doses of 4-MU was then assessed by a rat aortic ring assay. In addition, endometriotic lesions were induced in dorsal skinfold chambers of female BALB/c mice, which were daily treated with an intraperitoneal injection of 0.9% NaCl (vehicle group; n = 6), 20mg/kg 4-MU (n = 8) or 80mg/kg 4-MU (n = 7) throughout an observation period of 14 days. The effect of 4-MU on their vascularization, survival and growth were studied by intravital fluorescence microscopy, histology and immunohistochemistry. Main Results Non-cytotoxic doses of 4-MU effectively inhibited vascular sprout formation in the rat aortic ring assay. Endometriotic lesions in dorsal skinfold chambers of 4-MU-treated mice dose-dependently exhibited a significantly smaller vascularized area and lower functional microvessel density when compared to vehicle-treated controls. Histological analyses revealed a downregulation of HA expression in 4-MU-treated lesions. This was associated with a reduced density of CD31-positive microvessels within the lesions. In contrast, numbers of PCNA-positive proliferating and cleaved caspase-3-positive apoptotic cells did not differ between 4-MU-treated and control lesions. Conclusions The present study demonstrates for the first time that targeting the synthesis of HA suppresses angiogenesis in developing endometriotic lesions. Further studies have to clarify now whether in the future this anti-angiogenic effect can be used beneficially for the treatment of endometriosis.
Collapse
Affiliation(s)
- Carla N. Olivares
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
- * E-mail:
| | - Laura D. Alaniz
- CIT NOBA, Universidad Nacional del Noroeste de la Provincia de Buenos Aires (CONICET-UNNOBA), Junín, Buenos Aires, Argentina
| | - Michael D. Menger
- Institute for Clinical & Experimental Surgery, University of Saarland, Homburg/Saar, Germany
| | - Rosa I. Barañao
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Matthias W. Laschke
- Institute for Clinical & Experimental Surgery, University of Saarland, Homburg/Saar, Germany
| | - Gabriela F. Meresman
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
44
|
Marí-Alexandre J, Barceló-Molina M, Olcina-Guillem M, García-Oms J, Braza-Boïls A, Gilabert-Estellés J. MicroRNAs: New players in endometriosis. World J Obstet Gynecol 2016; 5:28-38. [DOI: 10.5317/wjog.v5.i1.28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/14/2015] [Accepted: 01/07/2016] [Indexed: 02/05/2023] Open
Abstract
Endometriosis is an estrogen-dependent inflammatory disorder that limits the quality of life of affected women. This pathology affects 10% of reproductive-age women, although the prevalence in those patients experiencing pain, infertility or both is as high as 35%-50%. Endometriosis is characterized by endometrial-like tissue outside the uterus, primarily on the pelvic peritoneum, ovaries and the pouch of Douglas. Despite extensive research endeavours, a unifying theory regarding the exact etiopathogenic mechanism of this high prevalent and incapacitating condition is still lacking, although it has been suggested that epigenetics could be involved. MicroRNAs (miRNAs), one of the epigenetic players, are small non-coding RNAs that can act as post-transcriptional regulators of gene expression, reducing the expression of their target mRNAs either inhibiting its translation or promoting its degradation. MiRNA expression profiles are specific of tissue and cell type. Abnormal miRNA expression has been described in different pathological conditions, such as a myriad of oncological, cardiovascular and inflammatory diseases and gynecological pathologies. In endometriosis, miRNA expression patterns of eutopic endometrium from patients and control women and from different endometriotic lesions have been described. These small non-coding molecules have become attractive candidates as novel biomarkers for an early non-invasive diagnosis of the disease, which could suppose a valuable benefit to the patients in terms of improvement of prognosis and reduction of the ratio of recurrence. In this systematic review we will focus on the role of miRNAs in the pathophisiology of endometriosis.
Collapse
|
45
|
Marí-Alexandre J, Sánchez-Izquierdo D, Gilabert-Estellés J, Barceló-Molina M, Braza-Boïls A, Sandoval J. miRNAs Regulation and Its Role as Biomarkers in Endometriosis. Int J Mol Sci 2016; 17:ijms17010093. [PMID: 26771608 PMCID: PMC4730335 DOI: 10.3390/ijms17010093] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/05/2016] [Accepted: 01/08/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs (18-22 nt) that function as modulators of gene expression. Since their discovery in 1993 in C. elegans, our knowledge about their biogenesis, function, and mechanism of action has increased enormously, especially in recent years, with the development of deep-sequencing technologies. New biogenesis pathways and sources of miRNAs are changing our concept about these molecules. The study of the miRNA contribution to pathological states is a field of great interest in research. Different groups have reported the implication of miRNAs in pathologies such as cancer, diabetes, cardiovascular, and gynecological diseases. It is also well-known that miRNAs are present in biofluids (plasma, serum, urine, semen, and menstrual blood) and have been proposed as ideal candidates as disease biomarkers. The goal of this review is to highlight the current knowledge in the field of miRNAs with a special emphasis to their role in endometriosis and the newest investigations addressing the use of miRNAs as biomarkers for this gynecological disease.
Collapse
Affiliation(s)
- Josep Marí-Alexandre
- Unit of Hemostasia, Thrombosis, Atherosclerosis and Vascular Biology, Health Research Institute La Fe, Valencia 46026, Spain.
| | | | | | - Moisés Barceló-Molina
- Unit of Hemostasia, Thrombosis, Atherosclerosis and Vascular Biology, Health Research Institute La Fe, Valencia 46026, Spain.
| | - Aitana Braza-Boïls
- Unit of Hemostasia, Thrombosis, Atherosclerosis and Vascular Biology, Health Research Institute La Fe, Valencia 46026, Spain.
| | - Juan Sandoval
- Epigomics Unit, Health Research Institute La Fe, Valencia 46026, Spain.
| |
Collapse
|
46
|
Expression of GRIM-19 in adenomyosis and its possible role in pathogenesis. Fertil Steril 2016; 105:1093-101. [PMID: 26769301 DOI: 10.1016/j.fertnstert.2015.12.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/16/2015] [Accepted: 12/16/2015] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To study the expression of the gene associated with retinoid-interferon (IFN)-induced mortality 19 (GRIM-19) in the endometrial tissue of patients with adenomyosis and to describe the possible pathogenic mechanisms of this phenomenon. DESIGN Experimental study using human samples and cell lines. SETTING University-affiliated hospital. PATIENT(S) Ectopic and eutopic endometrial tissues were obtained from 30 patients with adenomyosis, whereas normal endometrial specimens were obtained from 10 control patients without adenomyosis. INTERVENTION(S) Patients with rapid pathology report-confirmed adenomyosis were recruited, and eutopic and ectopic endometrial tissue samples were collected from patients who had undergone hysterectomies by either the transabdominal or laparoscopic method at Qilu Hospital. Normal endometrial tissue was collected from a group of control patients without adenomyosis. MAIN OUTCOME MEASURE(S) Immunohistochemistry (IHC) was performed to evaluate the expression of GRIM-19, phospho-signal transducer and activator of transcription 3 (Y705) (Y705) (pSTAT3(Y705)), and vascular endothelial growth factor (VEGF) in endometrial tissue samples. The protein levels of GRIM-19, pSTAT3(Y705), STAT3, and VEGF were detected by Western blot. Apoptosis in endometrial specimens was assayed by TUNEL. Immunohistochemistry with an antibody directed against CD34 was performed to detect new blood vessels in the endometrial tissue. GRIM-19 small interfering RNA and a recombinant plasmid carrying GRIM-19 were constructed to evaluate the effects of GRIM-19 on the downstream factors pSTAT3(Y705), STAT3, and VEGF in Ishikawa cells. RESULT(S) The expression of GRIM-19 was down-regulated in the eutopic endometria of patients with adenomyosis compared with the endometria of patients in the control group, and it was further reduced in the endometrial glandular epithelial cells of adenomyotic lesions. Apoptosis was reduced in the eutopic endometrium compared with the control group, and it was significantly reduced in ectopic endometrial tissues. In addition, the ectopic and eutopic endometria of patients with adenomyosis displayed a much higher microvessel density. In the eutopic and ectopic endometria of patients with adenomyosis, the expression levels of pSTAT3(Y705) and VEGF were significantly higher than in the controls. Furthermore, down-regulation of GRIM-19 in Ishikawa cells significantly promoted the activation of both pSTAT3(Y705) and its dependent gene VEGF. CONCLUSION(S) Aberrant expression of GRIM-19 may be associated with adenomyosis through the regulation of apoptosis and angiogenesis.
Collapse
|
47
|
Ultramicronized palmitoylethanolamide reduces viscerovisceral hyperalgesia in a rat model of endometriosis plus ureteral calculosis. Pain 2016; 157:80-91. [DOI: 10.1097/j.pain.0000000000000220] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
48
|
Vanhie A, Tomassetti C, Peeraer K, Meuleman C, D'Hooghe T. Challenges in the development of novel therapeutic strategies for treatment of endometriosis. Expert Opin Ther Targets 2015; 20:593-600. [PMID: 26558646 DOI: 10.1517/14728222.2016.1118461] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Endometriosis is an estrogen-dependent disease that results in pelvic pain and infertility. Its treatment is often frustrating due to limited medical treatment options, complex surgical treatment and high recurrence rates. Despite the advances in our understanding of the pathogenesis over the last decades and the consequent novel therapeutic strategies, no new drugs have been introduced in daily clinical practice. AREAS COVERED In the first part we present an overview of the pathogenesis of endometriosis. In the second part we discuss how new insights have led to the development of novel nonhormonal strategies for the treatment of endometriosis, focusing on anti-inflammatory and anti-angiogenic agents. In the third part we describe the problems encountered in the translation from experimental drugs to routine medicine for the treatment of endometriosis. EXPERT OPINION Despite the multitude of agents that have been tested in preclinical trials, only few drugs have passed to the stage of clinical testing and none have been introduced into clinical practice. It is our opinion that the major challenges in the translation from novel agents for endometriosis is due to the use of inadequate rodent models and a lack of standardization in the design and reporting of preclinical endometriosis models.
Collapse
Affiliation(s)
- Arne Vanhie
- a Leuven University Endometriosis Center of Expertise, Leuven University Fertility Center, Department of Obstetrics and Gynecology , UZ Gasthuisberg , Leuven , Belgium
| | - Carla Tomassetti
- a Leuven University Endometriosis Center of Expertise, Leuven University Fertility Center, Department of Obstetrics and Gynecology , UZ Gasthuisberg , Leuven , Belgium
| | - Karen Peeraer
- a Leuven University Endometriosis Center of Expertise, Leuven University Fertility Center, Department of Obstetrics and Gynecology , UZ Gasthuisberg , Leuven , Belgium
| | - Christel Meuleman
- a Leuven University Endometriosis Center of Expertise, Leuven University Fertility Center, Department of Obstetrics and Gynecology , UZ Gasthuisberg , Leuven , Belgium.,b Department of Gynecology and Obstetrics, Faculty of Medicine , Leuven University , Leuven , Belgium
| | - Thomas D'Hooghe
- b Department of Gynecology and Obstetrics, Faculty of Medicine , Leuven University , Leuven , Belgium
| |
Collapse
|
49
|
Santulli P, Marcellin L, Tosti C, Chouzenoux S, Cerles O, Borghese B, Batteux F, Chapron C. MAP kinases and the inflammatory signaling cascade as targets for the treatment of endometriosis? Expert Opin Ther Targets 2015; 19:1465-83. [PMID: 26389657 DOI: 10.1517/14728222.2015.1090974] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The pathogenesis of endometriosis, a common benign disease, remains ill-defined, although it is clear that chronic inflammation plays a crucial role through mitogen-activated protein kinase (MAPK) signaling pathways. All current medical therapies for endometriosis are antigonadotropic, and therefore have a contraceptive effect. A concerted research effort is hence warranted with the aim of delivering novel therapeutics that reduces disease symptoms without blocking ovulation. AREAS COVERED The authors review the complex pathogenic mechanisms of chronic inflammation in endometriosis and their relationships with MAPK pathways. The authors conducted a literature search of descriptive and functional targeted validation of MAPK in the pathogenesis of endometriosis. The effects of MAPK inhibitors, which constitute potential agents for future treatments, are also described. EXPERT OPINION Preliminary studies have highlighted a crucial role for MAPK in driving endometriosis-related inflammation. MAPK inhibitors exhibit potent activity in terms of controlling growth of endometriosis lesions both in vitro and in animal models. As MAPK inhibitors are known to have a multitude of undesirable side effects, their use in humans has to be approached with great care. Indeed, use of these drugs would probably be limited to short exposures prior to surgery in cases involving the most severe disease phenotypes.
Collapse
Affiliation(s)
- Pietro Santulli
- a 1 Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique - Hôpitaux de Paris (AP- HP), Groupe Hospitalier Universitaire (GHU) Ouest, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine , 75679 Paris, France +33 1 58 41 36 72 ; .,b 2 Université Paris Descartes, Sorbonne Paris Cité, Department "Development, Reproduction and Cancer," Institut Cochin , INSERM U1016, Equipe Pr Batteux, Paris, France
| | - Louis Marcellin
- a 1 Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique - Hôpitaux de Paris (AP- HP), Groupe Hospitalier Universitaire (GHU) Ouest, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine , 75679 Paris, France +33 1 58 41 36 72 ; .,b 2 Université Paris Descartes, Sorbonne Paris Cité, Department "Development, Reproduction and Cancer," Institut Cochin , INSERM U1016, Equipe Pr Batteux, Paris, France
| | - Claudia Tosti
- c 3 University of Siena, Obstetrics and Gynecology, Department of Molecular and Developmental Medicine , Siena, Italy
| | - Sandrine Chouzenoux
- b 2 Université Paris Descartes, Sorbonne Paris Cité, Department "Development, Reproduction and Cancer," Institut Cochin , INSERM U1016, Equipe Pr Batteux, Paris, France
| | - Olivier Cerles
- b 2 Université Paris Descartes, Sorbonne Paris Cité, Department "Development, Reproduction and Cancer," Institut Cochin , INSERM U1016, Equipe Pr Batteux, Paris, France
| | - Bruno Borghese
- a 1 Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique - Hôpitaux de Paris (AP- HP), Groupe Hospitalier Universitaire (GHU) Ouest, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine , 75679 Paris, France +33 1 58 41 36 72 ; .,b 2 Université Paris Descartes, Sorbonne Paris Cité, Department "Development, Reproduction and Cancer," Institut Cochin , INSERM U1016, Equipe Pr Batteux, Paris, France
| | - Frédéric Batteux
- b 2 Université Paris Descartes, Sorbonne Paris Cité, Department "Development, Reproduction and Cancer," Institut Cochin , INSERM U1016, Equipe Pr Batteux, Paris, France.,d 4 Hôpital Cochin, Department of Immunology , AP-HP, 75679 Paris cedex 14, France
| | - Charles Chapron
- a 1 Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique - Hôpitaux de Paris (AP- HP), Groupe Hospitalier Universitaire (GHU) Ouest, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine , 75679 Paris, France +33 1 58 41 36 72 ; .,b 2 Université Paris Descartes, Sorbonne Paris Cité, Department "Development, Reproduction and Cancer," Institut Cochin , INSERM U1016, Equipe Pr Batteux, Paris, France
| |
Collapse
|
50
|
Leconte M, Santulli P, Chouzenoux S, Marcellin L, Cerles O, Chapron C, Dousset B, Batteux F. Inhibition of MAPK and VEGFR by Sorafenib Controls the Progression of Endometriosis. Reprod Sci 2015; 22:1171-80. [PMID: 26169036 DOI: 10.1177/1933719115592708] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Sorafenib is a strong multikinase inhibitor targeting 2 different pathways of endometriosis pathogenesis: RAF kinase and vascular endothelial growth factor receptor (VEGFR). We investigate whether Sorafenib could control the growth of endometriotic lesions both in vitro and in vivo. METHODS Stromal primary cells were extracted from endometrial and endometriotic biopsies from patients with (n = 10) and without (n = 10) endometriosis. Proliferation, apoptosis, mitogen-activated protein kinases, and VEGFR-2 autophosphorylation were explored with and without Sorafenib treatment. Human endometriotic lesions were implanted in 30 nude mice randomized according to Sorafenib or placebo treatment. RESULTS Treating endometriotic cells with Sorafenib abrogated the phosphorylation of extracellular signal-regulated kinase in stromal cells of women with endometriosis compared to controls. In addition, this study highlights the antiangiogenic role of Sorafenib which translates as a decreased phosphorylated VEGFR-2-VEGFR-2 ratio in endometriosis. Using a xenogenic mouse model of endometriosis, we confirmed that Sorafenib regulates the endometriosis activity in vivo by targeting endometriosis-related proliferation and inflammation. CONCLUSION Our data suggest that Sorafenib controls the growth of endometriotic lesions in vitro and in vivo.
Collapse
Affiliation(s)
- Mahaut Leconte
- Department of "Development, Reproduction and Cancer", Institut Cochin, INSERM U1016, University Paris Descartes, Sorbonne Paris Cité, Paris, France Department of Digestive and Endocrine Surgery, Hôpital Cochin, AP-HP, Paris cedex, France
| | - Pietro Santulli
- Department of "Development, Reproduction and Cancer", Institut Cochin, INSERM U1016, University Paris Descartes, Sorbonne Paris Cité, Paris, France Department of Gynecology Obstetrics II and Reproductive Medicine, Faculty of Medicine, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Universitaire Ouest, Centre Hospitalier Universitaire Cochin, University Paris Descartes, Sorbonne Paris Cité, Paris, France Institut Cochin, University Paris Descartes, Sorbonne Paris Cité CNRS (UMR 8104), Paris, France
| | - Sandrine Chouzenoux
- Department of "Development, Reproduction and Cancer", Institut Cochin, INSERM U1016, University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Louis Marcellin
- Department of "Development, Reproduction and Cancer", Institut Cochin, INSERM U1016, University Paris Descartes, Sorbonne Paris Cité, Paris, France Department of Gynecology Obstetrics II and Reproductive Medicine, Faculty of Medicine, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Universitaire Ouest, Centre Hospitalier Universitaire Cochin, University Paris Descartes, Sorbonne Paris Cité, Paris, France Institut Cochin, University Paris Descartes, Sorbonne Paris Cité CNRS (UMR 8104), Paris, France
| | - Olivier Cerles
- Department of "Development, Reproduction and Cancer", Institut Cochin, INSERM U1016, University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Charles Chapron
- Department of "Development, Reproduction and Cancer", Institut Cochin, INSERM U1016, University Paris Descartes, Sorbonne Paris Cité, Paris, France Department of Gynecology Obstetrics II and Reproductive Medicine, Faculty of Medicine, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Universitaire Ouest, Centre Hospitalier Universitaire Cochin, University Paris Descartes, Sorbonne Paris Cité, Paris, France Institut Cochin, University Paris Descartes, Sorbonne Paris Cité CNRS (UMR 8104), Paris, France
| | - Bertrand Dousset
- Department of "Development, Reproduction and Cancer", Institut Cochin, INSERM U1016, University Paris Descartes, Sorbonne Paris Cité, Paris, France Department of Digestive and Endocrine Surgery, Hôpital Cochin, AP-HP, Paris cedex, France
| | - Frédéric Batteux
- Department of "Development, Reproduction and Cancer", Institut Cochin, INSERM U1016, University Paris Descartes, Sorbonne Paris Cité, Paris, France Department of Immunology, Hôpital Cochin, AP-HP, Paris cedex, France
| |
Collapse
|