1
|
Su H, Xie Z. Influencing factors and prediction model construction for recurrence in patients with ovarian endometriosis after laparoscopic conservative surgery. Am J Transl Res 2024; 16:7458-7466. [PMID: 39822533 PMCID: PMC11733349 DOI: 10.62347/yfce7581] [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: 09/12/2024] [Accepted: 11/11/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVE To investigate the factors influencing recurrence following laparoscopic conservative surgery in patients with ovarian endometriosis (OEM) and to develop a predictive model. METHODS In this retrospective study, the clinical data from 212 OEM patients who underwent laparoscopic conservative surgery at Suzhou Ninth People's Hospital from May 2013 to December 2021 were meticulously reviewed. According to disease recurrence over a 2-year follow-up period, the patients were divided into a recurrence group and a non-recurrence group. Univariate and multivariate logistic regression analyses were performed to identify factors associated with postoperative recurrence in OEM patients. A nomogram prediction model for postoperative recurrence in OEM patients was constructed using R 3.4.3 software. The discriminative power of the model was assessed using the area under the receiver operating characteristic (ROC) curve (AUC), with goodness of fit evaluated using the H-L goodness-of-fit test and Bootstrap method (self-sampling method). Clinical net benefit was analyzed through decision curve analysis. RESULTS Over a two-year follow-up, 36 cases of recurrence were observed, yielding a recurrence rate of 16.98%. Bilateral cysts (OR = 2.257, P = 0.005), high r-ASRM stage (OR = 2.651, P = 0.001), and elevated postoperative TNF-α levels (OR = 3.607, P = 0.004) were identified as risk factors for recurrence after laparoscopic conservative surgery in patients with OEM, while older age (OR = 0.566, P = 0.018) and postoperative adjuvant medication (OR = 0.509, P = 0.016) were protective factors. The nomogram prediction model, based on the above indicators, had an AUC of 0.895 for postoperative recurrence risk in OEM patients, with no overfitting phenomenon indicated by the goodness-of-fit test (χ2 = 1.786, P = 0.987). The Bootstrap validation (1000 samples) showed an average absolute error of 0.018 between predicted and actual probabilities. Decision curve analysis showed that the model effectively predicted a clinically relevant net benefit for postoperative recurrence risk. CONCLUSION A nomogram prediction model incorporating age, cyst distribution, r-ASRM staging, postoperative TNF-α levels, and postoperative adjuvant drugs effectively assesses the recurrence risk in OEM patients.
Collapse
Affiliation(s)
- Hailan Su
- Department of Gynecology, Suzhou Ninth People's Hospital Suzhou 215200, Jiangsu, China
| | - Zhijia Xie
- Department of Gynecology, Suzhou Ninth People's Hospital Suzhou 215200, Jiangsu, China
| |
Collapse
|
2
|
Rencber SF, Yazır Y, Sarıhan M, Sezer Z, Korun ZEU, Ozturk A, Duruksu G, Guzel E, Akpınar G, Corakci A. Endoplasmic reticulum stress of endometrial mesenchymal stem cells in endometriosis. Tissue Cell 2024; 91:102544. [PMID: 39217786 DOI: 10.1016/j.tice.2024.102544] [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: 07/22/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE The human endometrium has significant regenerative abilities due to stem cells, which are vital in immunomodulation, immune tolerance, steroid hormone response, and inflammation. Endometriosis, an inflammatory gynecological disorder where endometrium-like tissue grows outside uterus, affects millions of women and often causes infertility. Recent research indicates that stem cells contribute to pathology of endometriosis. ER stress is implicated in various diseases, including endometriosis. This study aims to examine ER stress in eMSCs within endometriosis pathogenesis and uncover underlying disease mechanisms. METHODS Samples were collected from healthy subjects and women with endometriosis in both proliferative and secretory phases. eMSCs were isolated and characterized via flow cytometry. ER stress protein levels were assessed using proteomic analysis, with validation through Western Blot and immunofluorescence staining. Gene expression was analyzed by RT-qPCR, and ultrastructural examination of eMSCs was conducted using TEM. ER stress markers in tissue samples were detected in SUSD2+ eMSCs through immunofluorescence staining and visualized using a confocal microscope. Statistical analysis was performed using SPSS program. RESULTS The proteomics analysis uncovered ER stress-related proteins (DDRGK1, RTN3, ERp44, TMED2, TMEM33, TMX3) whose levels were significantly distinct from control group. Western Blot analysis and immunofluorescence staining results at protein level; RT-qPCR results at gene level supported these findings. TEM analysis also showed ultrastructural presence of ER stress in endometriosis groups. CONCLUSION Presence of ER stress in eMSCs in pathogenesis of endometriosis has been demonstrated using various methods. Our research has potential to shed light on pathology of endometriosis and offer promising avenues for non-invasive diagnosis and potential treatment.
Collapse
Affiliation(s)
- Selenay Furat Rencber
- Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Yusufhan Yazır
- Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey.
| | - Mehmet Sarıhan
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Zehra Sezer
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeynep Ece Utkan Korun
- Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Department of Obstetrics and Gynecology, Faculty of Medicine, Yeditepe University, İstanbul, Turkey
| | - Ahmet Ozturk
- Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Gokhan Duruksu
- Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Elif Guzel
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gurler Akpınar
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Aydın Corakci
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
3
|
Cho SB. Molecular Mechanisms of Endometriosis Revealed Using Omics Data. Biomedicines 2023; 11:2210. [PMID: 37626707 PMCID: PMC10452455 DOI: 10.3390/biomedicines11082210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/22/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Endometriosis is a gynecological disorder prevalent in women of reproductive age. The primary symptoms include dysmenorrhea, irregular menstruation, and infertility. However, the pathogenesis of endometriosis remains unclear. With the advent of high-throughput technologies, various omics experiments have been conducted to identify genes related to the pathophysiology of endometriosis. This review highlights the molecular mechanisms underlying endometriosis using omics. When genes identified in omics experiments were compared with endometriosis disease genes identified in independent studies, the number of overlapping genes was moderate. However, the characteristics of these genes were found to be equivalent when functional gene set enrichment analysis was performed using gene ontology and biological pathway information. These findings indicate that omics technology provides invaluable information regarding the pathophysiology of endometriosis. Moreover, the functional characteristics revealed using enrichment analysis provide important clues for discovering endometriosis disease genes in future research.
Collapse
Affiliation(s)
- Seong Beom Cho
- Department of Biomedical Informatics, College of Medicine, Gachon University, 38-13, Dokgeom-ro 3 Street Namdon-gu, Incheon 21565, Republic of Korea
| |
Collapse
|
4
|
Martone S, Troìa L, Marcolongo P, Luisi S. Role of medical treatment of endometriosis. Minerva Obstet Gynecol 2021; 73:304-316. [PMID: 34008385 DOI: 10.23736/s2724-606x.21.04784-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Endometriosis is a chronic benign disease that affects women of reproductive age. Medical therapy is often the first line of management for women with endometriosis in order to ameliorate symptoms or to prevent post-surgical disease recurrence. Currently, there are several medical options for the management of patients with endometriosis and long-term treatments should balance clinical efficacy (controlling pain symptoms and preventing recurrence of disease after surgery) with an acceptable safety-profile. Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used in the treatment of chronic inflammatory conditions, being efficacious in relieving primary dysmenorrhea. Combined oral contraceptives and progestins, available for multiple routes of administration, are commonly administered as first-line hormonal therapies. Several studies demonstrated that they succeed in improving pain symptoms in the majority of patients; moreover, they are well tolerated and not expensive. Gonadotropin-releasing hormone-agonists are prescribed when first line therapies are ineffective, not tolerated or contraindicated. Even if these drugs are efficacious in treating women not responding to COCs or progestins, they are not orally available and have a less favorable tolerability profile (needing an appropriate add-back therapy). Because few data are available on long-term efficacy and safety of aromatase inhibitors they should be reserved only for women with symptoms who are refractory to other treatments only in a research environment. Almost all of the currently available treatment options for endometriosis suppress ovarian function and are not curative. For this reason, research into new drugs is unsurprisingly demanding. Amongst the drugs currently under investigation, gonadotropin-releasing hormone antagonists have shown most promise, currently in late-stage clinical development. There is a number of potential future therapies currently tested only in vitro, in animal models of endometriosis or in early clinical studies with a small sample size. Further studies are necessary to conclude whether these treatments would be of value for the treatment of endometriosis.
Collapse
Affiliation(s)
- Simona Martone
- Department of Molecular and Developmental Medicine, Obstetrics and Gynecology, University of Siena, Siena, Italy
| | - Libera Troìa
- Department of Molecular and Developmental Medicine, Obstetrics and Gynecology, University of Siena, Siena, Italy
| | - Paola Marcolongo
- Department of Molecular and Developmental Medicine, Obstetrics and Gynecology, University of Siena, Siena, Italy
| | - Stefano Luisi
- Department of Molecular and Developmental Medicine, Obstetrics and Gynecology, University of Siena, Siena, Italy -
| |
Collapse
|
5
|
Kocbek V, Imboden S, Nirgianakis K, Mueller M, McKinnon B. Dual influence of TNFα on diverse in vitro models of ovarian cancer subtypes. Heliyon 2021; 7:e06099. [PMID: 33615005 PMCID: PMC7881225 DOI: 10.1016/j.heliyon.2021.e06099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 01/03/2021] [Accepted: 01/22/2021] [Indexed: 12/05/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological cancer. Numerous subtypes exist, each with distinct risk factors and prognosis. What underlies these subtypes and their progression is not clear, although inflammation through NFκB may play a key role. We performed a study on a series of well-characterized in vitro ovarian cancer models including TOV21G, TOV112D and OV90 originally derived from clear cell, endometrioid and high grade serous carcinoma respectively. Cells were treated with 0-100 ng/ml TNFα over 6-72 h. The NFκB pathway was inhibited by a series of NFκB pathway inhibitors, 100μM PDTC, 1μM PS-1145 and 200nM TPCA and the influence on cellular viability and inflammation was measured via an MTS assay and qPCR respectively. TNFα stimulation of NFκB was confirmed via Western blot. We found TNFα facilitated continued growth of TOV21G and TOV112D cells in an NFκB independent method. In contrast, TNFα inhibited OV90 cell growth in an NFκB dependent manner. TNFα stimulated production of IL-6, IL-8, MCP-1 and RANTES on all three cells lines, but only IL-6 and IL-8 were via NFκB mediated mechanisms. These results indicate TNFα may have diverse effects mediated through both NFκB and non-NFκB pathways on ovarian cancer cells. Understanding the role for TNFα in each subtype may have significant implications for charting disease progression and designing personalized treatments.
Collapse
Affiliation(s)
- Vida Kocbek
- Department of Clinical Research, University of Berne, Murtenstrasse 35, Berne, CH-3010, Switzerland
- Department of Obstetrics and Gynaecology, Inselspital, Berne University Hospital, Effingerstrasse 102, Berne, CH-3010, Switzerland
| | - Sara Imboden
- Department of Clinical Research, University of Berne, Murtenstrasse 35, Berne, CH-3010, Switzerland
- Department of Obstetrics and Gynaecology, Inselspital, Berne University Hospital, Effingerstrasse 102, Berne, CH-3010, Switzerland
| | - Kostantinos Nirgianakis
- Department of Clinical Research, University of Berne, Murtenstrasse 35, Berne, CH-3010, Switzerland
- Department of Obstetrics and Gynaecology, Inselspital, Berne University Hospital, Effingerstrasse 102, Berne, CH-3010, Switzerland
| | - Michael Mueller
- Department of Clinical Research, University of Berne, Murtenstrasse 35, Berne, CH-3010, Switzerland
- Department of Obstetrics and Gynaecology, Inselspital, Berne University Hospital, Effingerstrasse 102, Berne, CH-3010, Switzerland
| | - Brett McKinnon
- Department of Obstetrics and Gynaecology, Inselspital, Berne University Hospital, Effingerstrasse 102, Berne, CH-3010, Switzerland
| |
Collapse
|
6
|
Liu J, Wen S, Lin Y, Yang X, Liu Z, Quan S, Song Y. Advanced oxidation protein products change biological behaviors of rat endometrial epithelial cells by activating ERK/P38 signaling pathways. Biol Open 2020; 9:bio048876. [PMID: 32366372 PMCID: PMC7286297 DOI: 10.1242/bio.048876] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 04/17/2020] [Indexed: 12/31/2022] Open
Abstract
Advanced oxidation protein products (AOPPs) are a family of oxidized protein compounds and could induce oxidative stress and inflammatory lesion in various cells. The accumulation of AOPPs was associated with female reproductive diseases such as polycystic ovary syndrome (PCOS), leiomyoma and endometriosis. However, the relationship between AOPPs and endometrial cells is unclear. To explore the effects of accumulated AOPPs on endometrial cells, we treated normal rat endometrial epithelial cells (rEECs) and endometriosis model rats with AOPPs. Primary rEECs were collected from 8-week-old female Wistar rats. Increasing the amount of AOPPs in the media of rEECs enhanced rEEC proliferation and migration, and inhibited apoptosis. Moreover, AOPPs triggered the production of reactive oxygen species and nitrite along with activated ERK and P38 signal and this, in turn, led to an upregulation of proliferation and migration. With the treatment of antioxidants or the inhibitors of ERK and P38, the above effects of AOPPs on rEECs were attenuated. Additionally, in an endometriosis rat model, a similar phenomenon was observed in that the growth of endometriotic implants were promoted by AOPPs and EECs were significantly increased. This study indicated that the accumulation of AOPPs could promote rEEC proliferation and migration through ERK and P38 signal both in vivo and in vitroThis article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jing Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Sixi Wen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen 518000, People's Republic of China
| | - Yanling Lin
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Xiaoping Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Zebang Liu
- College of Engineering, Huazhong Agricultural University, Wuhan 43000, People's Republic of China
| | - Song Quan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yali Song
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| |
Collapse
|
7
|
Tamaddoni A, Mohammadi E, Sedaghat F, Qujeq D, As'Habi A. The anticancer effects of curcumin via targeting the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway. Pharmacol Res 2020; 156:104798. [PMID: 32278045 DOI: 10.1016/j.phrs.2020.104798] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 01/19/2023]
Abstract
The mammalian target of rapamycin (mTOR) is a protein kinase that has been considered as a key regulator of a large number of cellular processes, including cell growth, proliferation, differentiation, survival, and motility. Overactivation of mTOR (especially mTORC1) signaling is related to oncogenic cellular processes. Therefore targeting mTORC1 signaling is a new promising strategy in cancer therapy. In this regard, various studies have shown that curcumin, a polyphenol produced from the turmeric rhizome, has anti-inflammatory, antioxidant and anticancer properties. Curcumin may exert its anticancer function, at least in part, by suppressing mTOR-mediated signaling pathway in tumor cells. However, the exact underlying mechanisms by which curcumin blocks the mTORC1 signaling remain unclear. According to literature, curcumin inhibits insulin-like growth factor 1 (IGF-1)/phosphoinositide 3-kinase (PI3K)/Akt/mTORC1 pathway which leads to apoptosis and cell cycle arrest via suppression of erythroblastosis virus transcription factor 2 and murine double minute 2 oncoprotein. In addition, activation of unc-51-like kinase 1 by curcumin, as a downstream target of IGF-1/PI3K/Akt/mTORC1 axis, enhances autophagy. Curcumin induces AMP-activated protein kinase, a negative regulator of mTORC1, via inhibition of F0F1-ATPase. Interestingly, curcumin suppresses IκB kinase β, the upstream kinase in mTORC1 pathway. Moreover, evidence revealed that curcumin downregulates the E3-ubiquitin ligases NEDD4, neural precursor cell-expressed developmentally downregulated 4. NEDD4 is frequently overexpressed in a wide range of cancers and degrades the phosphatase and tensin homolog, which is a negative regulator of mTORC1. Finally another suggested mechanism is suppression of MAOA/mTORC1/hypoxia-inducible factor 1α signaling pathway by curcumin.
Collapse
Affiliation(s)
- Ahmad Tamaddoni
- Non-Communicable Pediatric Diseases Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Elahe Mohammadi
- Department of Nutrition, Khalkhal University of Medical Sciences, Khalkhal, Iran.
| | - Fatemeh Sedaghat
- Department of Basic Medical Sciences, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Atefeh As'Habi
- Food Safety Research Center (Salt), Semnan University of Medical Sciences, Semnan, Iran; Department of Nutrition, School of Nutrition and Food Sciences, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
8
|
Qi H, Zhang H, Zhao X, Qin Y, Liang G, He X, Zhang J. Integrated analysis of mRNA and protein expression profiling in tubal endometriosis. Reproduction 2020; 159:601-614. [PMID: 32130204 PMCID: PMC7159149 DOI: 10.1530/rep-19-0587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
Tubal endometriosis (tubal EM) is a subtype of endometriosis (EM) associated with fallopian tube impairments and infertility. Since the molecular mechanism underlying tubal EM is not clear, we assume that an aberrant transcriptome of fallopian tube epithelium and microenvironment changes caused by cytokines in tubal fluid are possible causes. The aim of this study was to identify potential hub mRNAs/proteins of tubal EM through integrated transcriptomic and proteomic analyses and to elucidate significant pathways, cellular functions, and interaction networks during the initiation and progression of tubal EM. We obtained human fallopian tube epithelium and tubal fluid samples from patients with and without tubal EM. Tubal epithelia were analyzed using microarray, and tubal fluid was analyzed using quantitative label-free LC-MS/MS. We identified differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) and determined common mRNAs/protein. We observed 35 commonly deregulated mRNAs/proteins, and IPA indicated that cellular movement, inflammatory response, and immune cell trafficking were significantly activated during the pathogenesis of tubal EM. We also identified acute phase response signaling pathway activation as a unique pathogenesis signature of tubal EM. Our results demonstrate that an integrated analysis of the transcriptome and proteome has the potential to reveal novel disease mechanisms at a molecular level.
Collapse
Affiliation(s)
- Hang Qi
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Huiyu Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Xiaoya Zhao
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Ya Qin
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Guiling Liang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Xiaoqing He
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Jian Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| |
Collapse
|
9
|
Di Nisio V, Rossi G, Di Luigi G, Palumbo P, D'Alfonso A, Iorio R, Cecconi S. Increased levels of proapoptotic markers in normal ovarian cortex surrounding small endometriotic cysts. Reprod Biol 2019; 19:225-229. [PMID: 31416694 DOI: 10.1016/j.repbio.2019.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 02/07/2023]
Abstract
Endometriosis can impair fertility by reducing ovarian reserve and the production of good-quality oocytes. The surgical removal of endometriotic lesions is generally recommended for women who wish to conceive. In this paper we studied whether ovarian cortex adjacent to excised small (diameter ≤ 4 cm) endometriotic cyst (here referred as Cortex Surrounding Endometriotic Cyst, CSEC) showed signs of tissue damages by evaluating the expression of proteins involved in DNA repair and apoptosis. To this end, phosphorylated H2A.X, Chk1 and 2, ATM and ATR, Bcl-2, Bid, phosphorylated and total p53, caspases (9, 8 and 3), XIAP, phosphorylated and total NFκB were analyzed by western blot. Results showed that caspase 8, XIAP, p53/p-p53 and NFκB were more abundantly expressed in all samples of CSEC group in comparison with ovarian cortex of controls. Conversely, the levels of the other proteins were comparable between the two groups. In conclusion, these results suggest that NFκB, caspase 8 and p53/p-p53 elevated expressions in samples of CSEC can be considered as an early sign of tissue injury, indicating that ovarian cortex is already sensitized to apoptosis and inflammation. Therefore, excision of EC should occur very early, to avoid further ovarian damages.
Collapse
Affiliation(s)
- Valentina Di Nisio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Gianna Rossi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Paola Palumbo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Angela D'Alfonso
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Roberto Iorio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Sandra Cecconi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
10
|
Ameliorative effects of luteolin against endometriosis progression in vitro and in vivo. J Nutr Biochem 2019; 67:161-172. [PMID: 30925413 DOI: 10.1016/j.jnutbio.2019.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/21/2019] [Accepted: 02/25/2019] [Indexed: 12/24/2022]
Abstract
Endometriosis is a common gynecological disease in reproductive-aged women. Generally, accumulation of backflow and debris of endometrial tissue develops into a lesion outside of the endometrium, inducing severe pelvic pain and infertility in some patients. Hormone therapy and surgery are the main treatments available, but various therapeutic phytochemicals are being reviewed in animal studies or clinical trials for endometriosis patients nowadays. However, the therapeutic effects of luteolin in human endometriosis have not been studied well. Here, we demonstrate that luteolin exerts antiproliferative and apoptotic effects in human VK2/E6E7 and End1/E6E7 and in an animal endometriosis model. Luteolin inhibits cell proliferation through cell cycle arrest and induces apoptosis through DNA fragmentation in VK2/E6E7 and End1/E6E7 cells. Cytosolic calcium levels, ROS production and lipid peroxidation also increased dose-dependently (0, 5, 10 and 20 μM) in the treatment with luteolin. In VK2/E6E7 and End1/E6E7 cells, luteolin decreased ERK1/2, JNK and PI3K/AKT signal proteins while activating P38. In addition, intraperitoneal injection of luteolin in the endometriosis mouse model reduced lesion size compared to vehicle-injected mice. Ccne1, Cdk2 and Cdk4 were significantly down-regulated in the autoimplanted endometriosis lesions of mice intraperitoneally injected with luteolin. Knockdown of CCNE1 mRNA in VK2/E6E7 and End1/E6E7 cells decreased cell viability through inhibition of G0/G1 phase progression and increased apoptosis. Together, our results imply that luteolin suppresses endometriosis development by regulation of the PI3K/AKT and MAPK signal proteins as well as the expression of CCNE1 in vitro and in vivo.
Collapse
|
11
|
McKinnon B, Mueller M, Montgomery G. Progesterone Resistance in Endometriosis: an Acquired Property? Trends Endocrinol Metab 2018; 29:535-548. [PMID: 29934050 DOI: 10.1016/j.tem.2018.05.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 12/21/2022]
Abstract
Endometriosis is the growth of endometrial tissue outside the uterus and is characterized by progesterone resistance and changes in global and progesterone target gene expression. However, the mechanism behind this and whether it is innate, acquired, or present in both the eutopic and ectopic tissue in not always clear. We find large-scale gene expression studies in eutopic tissue, indicative of progesterone resistance, are often contradictory, potentially due to the dynamic nature of this tissue, whereas suppressed progesterone receptor expression is supported in ectopic but not eutopic tissue. This suggests more studies are required in eutopic tissue particularly, and that potentially the suppressed progesterone receptor (PR) expression is a consequence of the pathogenic process and exposure to the peritoneal environment.
Collapse
Affiliation(s)
- Brett McKinnon
- Department of Gynecology and Obstetrics, Frauenklinik, Inselspital Bern, Switzerland.
| | - Michael Mueller
- Department of Gynecology and Obstetrics, Frauenklinik, Inselspital Bern, Switzerland
| | - Grant Montgomery
- Genomics of Reproductive Disorders, Institute for Molecular Bioscience, University of Queensland, Australia
| |
Collapse
|
12
|
Zou Y, Zhou JY, Wang F, Zhang ZY, Liu FY, Luo Y, Tan J, Zeng X, Wan XD, Huang OP. Analysis of CARD10 and CARD11 somatic mutations in patients with ovarian endometriosis. Oncol Lett 2018; 16:491-496. [PMID: 29928437 DOI: 10.3892/ol.2018.8659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/27/2018] [Indexed: 12/17/2022] Open
Abstract
Endometriosis is a complex and heterogeneous pre-malignant inflammatory disease harboring multiple gene mutations. Previous studies have suggested that caspase recruitment domain family member (CARD)10 and CARD11 mutations may exist in endometriosis. In the present study, a collection of endometriotic lesions and paired peripheral blood from 101 patients with ovarian endometriosis were obtained, and the entire coding sequences of the CARD10 and CARD11 genes were sequenced. Evolutionary conservation analysis and online prediction programs were applied to analyze the disease-causing potential of the identified mutations. A total of 4 novel somatic mutations were identified in 4 out of the 101 (4.0%) samples: 2 in-frame deletions in CARD10 (c.785_790delAGGAGA, p.K272_E273delKE; c.785_802delAGGAGAAGGAGAAGGAGA, p.K272_V277delKEPDNV) and 2 heterozygous missense mutations in CARD11 (c.49G>T, p.D17Y; c.160G>C, p.E54Q). The sample with CARD10 p.K272_E273delKE deletion was obtained from a 47-year-old patient who was also diagnosed with uterine leiomyoma, while the CARD10 p.K272_V277delKEPDNV-mutated sample was from a 43-year-old patient exhibiting a decreased blood eosinophil granulocyte ratio (0.3%) and an elevated serum creatine kinase level (314 U/l). The patient with the CARD11 p.D17Y mutation was 38 years old and exhibited an increased level of cancer antigen 125 (45.4 U/ml), while the patient with the CARD11 p.E54Q mutation was 46 years old and exhibited no other gynecological conditions. Evolutionary conservation analysis and online prediction programs suggested that these mutations may be disease-causing. In summary, 4 novel somatic mutations in the CARD10 and CARD11 genes were identified from amongst 101 cases of ovarian endometriosis for the first time, these mutations may serve active roles in the development of ovarian endometriosis.
Collapse
Affiliation(s)
- Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Jiang-Yan Zhou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Feng Wang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Zi-Yu Zhang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Fa-Ying Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Luo
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Jun Tan
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Reproductive Medicine Center, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Xin Zeng
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Xi-Di Wan
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Ou-Ping Huang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
13
|
Sbracia M, McKinnon B, Scarpellini F, Marconi D, Rossi G, Simmilion C, Mueller MD, Barnea ER, Mueller M. PreImplantation Factor in endometriosis: A potential role in inducing immune privilege for ectopic endometrium. PLoS One 2017; 12:e0184399. [PMID: 28902871 PMCID: PMC5597204 DOI: 10.1371/journal.pone.0184399] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 08/23/2017] [Indexed: 01/19/2023] Open
Abstract
Endometriosis is a chronic inflammatory condition characterised by the growth of endometrial epithelial and stromal cells outside the uterine cavity. In addition to Sampson’s theory of retrograde menstruation, endometriosis pathogenesis is facilitated by a privileged inflammatory microenvironment, with T regulatory FoxP3+ expressing T cells (Tregs) being a significant factor. PreImplantation Factor (PIF) is a peptide essential for pregnancy recognition and development. An immune modulatory function of the synthetic PIF analog (sPIF) has been successfully confirmed in multiple animal models. We report that PIF is expressed in the epithelial ectopic cells in close proximity to FoxP3+ stromal cells. We provide evidence that PIF interacts with FoxP3+ cells and modulates cell viability, dependent on cell source and presence of inflammatory mediators. Our finding represent a novel PIF-based mechanism in endometriosis that has potential for novel therapeutics.
Collapse
Affiliation(s)
- Marco Sbracia
- Hungaria Center for Endocrinology and Reproductive Medicine, Rome, Italy
| | - Brett McKinnon
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Fabio Scarpellini
- Hungaria Center for Endocrinology and Reproductive Medicine, Rome, Italy
| | - Daniela Marconi
- Department of Obstetrics and Gynecology, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Gabriele Rossi
- Department of Obstetrics and Gynecology, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Cedric Simmilion
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Michael D. Mueller
- Department of Clinical Research, University of Bern, Bern, Switzerland
- Department of Obstetrics and Gynecology, University Hospital Bern, Bern, Switzerland
| | - Eytan R. Barnea
- SIEP- The Society for the Investigation of Early Pregnancy, Cherry Hill, NJ, United States of America
- Department of Research and Development, BioIncept LLC, Cherry Hill, NJ, United States of America
- * E-mail: (MM); (ERB)
| | - Martin Mueller
- Department of Clinical Research, University of Bern, Bern, Switzerland
- Department of Obstetrics and Gynecology, University Hospital Bern, Bern, Switzerland
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, United States of America
- * E-mail: (MM); (ERB)
| |
Collapse
|
14
|
Long-term medical management of endometriosis with dienogest and with a gonadotropin-releasing hormone agonist and add-back hormone therapy. Fertil Steril 2017; 107:537-548. [DOI: 10.1016/j.fertnstert.2016.12.024] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/19/2016] [Accepted: 12/22/2016] [Indexed: 11/22/2022]
|