1
|
Antonis S, Efthymia P, Maria K, Eleftherios P, Sotiris K, Neoklis G, Fuminori T, George A, Tasuku H, Apostolos K. The effect of combined oral contraceptive pills on angiogenesis in endometriotic lesions. Hormones (Athens) 2025:10.1007/s42000-025-00636-4. [PMID: 39982662 DOI: 10.1007/s42000-025-00636-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
PURPOSE Neoangiogenesis is necessary for adhesion and invasion of endometriotic lesions. We hypothesize that by blocking angiogenetic pathways we can suppress endometriosis. Oral contraceptive pills (OCs) are routinely used in endometriosis to suppress symptoms of the disease. In the current study, we attempt to evaluate the effects of OCs on various angiogenetic factors in women with endometriosis. METHODS Sixty women with endometriosis were randomly divided into two groups. Group A consisted of 30 women who received OCs in a cyclical manner for 3 months before surgery and group B of 30 women who did not. Biopsy specimens of ovarian endometrioma were collected. We used qRT-PCR to study the mRNA expression levels of VEGF, TF, PAR-2, SP1, and FGF1. RESULTS The levels of mRNA of all angiogenic factors were found to be elevated in women who received OCs compared with women who did not. This difference was statistically significant for VEGF, TF, FGF1, SP1 (p < 0.001), and PAR-2 (p = 0.046). CONCLUSION OC administration does not inhibit neoangiogenesis in endometriotic lesions; on the contrary, angiogenetic pathways might be upregulated.
Collapse
Affiliation(s)
- Siampalis Antonis
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | | | - Keramida Maria
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Panteris Eleftherios
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | | | - Georgopoulos Neoklis
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Taniguchi Fuminori
- Dept. of Obstetrics & Gynecology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Adonakis George
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Harada Tasuku
- Dept. of Obstetrics & Gynecology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kaponis Apostolos
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece.
| |
Collapse
|
2
|
Filindris T, Papakonstantinou E, Keramida M, Panteris E, Kalogeropoulos S, Georgopoulos N, Taniguchi F, Adonakis G, Harada T, Kaponis A. The effect of GnRH-a on the angiogenesis of endometriosis. Hormones (Athens) 2024; 23:509-515. [PMID: 38639888 PMCID: PMC11436414 DOI: 10.1007/s42000-024-00559-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
PURPOSE Neoangiogenesis is necessary for adhesion and invasiveness of endometriotic lesions in women affected by endometriosis. Vascular endothelial growth factor (VEGF) is one of the main components of angiogenesis and is part of the major pathway tissue factor (TF)-protease activated receptor-2 (PAR-2)-VEGF that leads to neoangiogenesis. Specificity protein 1 (SP1) is a transcriptional factor that has recently been studied for its crucial role in angiogenesis via a specific pathway. We hypothesize that by blocking angiogenetic pathways we can suppress endometriotic lesions. Gonadotrophin-releasing hormone-agonists (GnRH-a) are routinely used, especially preoperatively, in endometriosis. It would be of great interest to clarify which angiogenetic pathways are affected and, thereby, pave the way for further research into antiangiogenetic effects on endometriosis. METHODS We used quantitative real-time polymerase chain reaction (qRT-PCR) to study mRNA expression levels of TF, PAR-2, VEGF, and SP1 in endometriotic tissues of women who underwent surgery for endometriosis and received GnRH-a (leuprolide acetate) preoperatively. RESULTS VEGF, TF, and PAR-2 expression is significantly lower in patients who received treatment (p < 0,001) compared to those who did not, whereas SP1 expression is not altered (p = 0.779). CONCLUSIONS GnRH-a administration does affect some pathways of angiogenesis in endometriotic lesions, but not all of them. Therefore, supplementary treatments that affect the SP1 pathway of angiogenesis should be developed to enhance the antiangiogenetic effect of GnRH-a in patients with endometriosis. TRIAL REGISTRATION Clinicaltrial.gov ID: NCT06106932.
Collapse
Affiliation(s)
- Theodoros Filindris
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | | | - Maria Keramida
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Eleftherios Panteris
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | | | - Neoklis Georgopoulos
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Fuminori Taniguchi
- Dept. of Obstetrics & Gynecology, Tottori University Faculty of Medicine, Yonago, Japan
| | - George Adonakis
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece
| | - Tasuku Harada
- Dept. of Obstetrics & Gynecology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Apostolos Kaponis
- Dept. of Obstetrics & Gynecology, Patras University School of Medicine, Patras, Greece.
| |
Collapse
|
3
|
Zaninelli TH, Fattori V, Heintz OK, Wright KR, Bennallack PR, Sim D, Bukhari H, Terry KL, Vitonis AF, Missmer SA, Andrello AC, Anchan RM, Godin SK, Bree D, Verri WA, Rogers MS. Targeting NGF but not VEGFR1 or BDNF signaling reduces endometriosis-associated pain in mice. J Adv Res 2024:S2090-1232(24)00360-6. [PMID: 39142441 DOI: 10.1016/j.jare.2024.08.017] [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: 12/13/2023] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024] Open
Abstract
INTRODUCTION Endometriosis is a chronic inflammatory disease that affects ∼10 % of women. A significant fraction of patients experience limited or no efficacy with current therapies. Tissue adjacent to endometriosis lesions often exhibits increased neurite and vascular density, suggesting that disease pathology involves neurotrophic activity and angiogenesis. OBJECTIVES We aim to evaluate the potential for key tyrosine-kinase-receptor-coupled neurotrophic molecules to contribute to endometriosis-associated pain in mice. METHODS Peritoneal fluid was collected from endometriosis patients undergoing surgery and the levels of NGF and VEGFR1 regulators (VEGFA, VEGFB, PLGF, and sVEGFR1) were quantified by ELISA. VEGFR1 regulator concentrations were used to calculate VEGFR1 occupancy. We used genetic depletion, neutralizing antibodies, and pharmacological approaches to specifically block neurotrophic ligands (NGF or BDNF) or receptors (VEGFR1, TRKs) in a murine model of endometriosis-associated pain. Endometriosis-associated pain was measured using von Frey filaments, quantification of spontaneous abdominal pain-related behavior, and thermal discomfort. Disease parameters were evaluated by lesion size and prevalence. To evaluate potential toxicity, we measured the effect of entrectinib dose and schedule on body weight, liver and kidney function, and bone structure (via micro-CT). RESULTS We found that entrectinib (pan-Trk inhibitor) or anti-NGF treatments reduced evoked pain, spontaneous pain, and thermal discomfort. In contrast, even though calculated receptor occupancy revealed that VEGFR1 agonist levels are sufficient to support signaling, blocking VEGFR1 via antibody or tamoxifen-induced knockout did not reduce pain or lesion size in mice. Targeting BDNF-TrkB with an anti-BDNF antibody also proved ineffective. Notably, changing dosing schedule to once weekly eliminated entrectinib-induced bone-loss without decreasing efficacy against pain. CONCLUSIONS This suggests NGF-TrkA signaling, but not BDNF-TrkB or VEGF-VEGFR1, mediates endometriosis-associated pain. Moreover, entrectinib blocks endometriosis-associated pain and reduces lesion sizes. Our results also indicated that entrectinib-like molecules are promising candidates for endometriosis treatment.
Collapse
Affiliation(s)
- Tiago H Zaninelli
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States; Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, PR, Brazil
| | - Victor Fattori
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Olivia K Heintz
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kristeena R Wright
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Philip R Bennallack
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Danielle Sim
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Hussain Bukhari
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kathryn L Terry
- Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Boston Center for Endometriosis, Boston Children's Hospital and Brigham and Women's Hospital, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Allison F Vitonis
- Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Boston Center for Endometriosis, Boston Children's Hospital and Brigham and Women's Hospital, Boston, MA, United States
| | - Stacey A Missmer
- Boston Center for Endometriosis, Boston Children's Hospital and Brigham and Women's Hospital, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Obstetrics, Gynecology, and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Avacir C Andrello
- Department of Physics, Center of Exact Sciences, Londrina State University, Londrina, PR, Brazil
| | - Raymond M Anchan
- Boston Center for Endometriosis, Boston Children's Hospital and Brigham and Women's Hospital, Boston, MA, United States; Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Dara Bree
- Cygnal Therapeutics, Cambridge, MA, United States
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, PR, Brazil
| | - Michael S Rogers
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States; Boston Center for Endometriosis, Boston Children's Hospital and Brigham and Women's Hospital, Boston, MA, United States.
| |
Collapse
|
4
|
Shi J, Jing W, He Y, Huang Y. Decreased expression of KLF6 in ectopic endometrial stromal cells contributes to endometriosis progression by targeting CTNNB1. Cell Signal 2024; 120:111230. [PMID: 38761988 DOI: 10.1016/j.cellsig.2024.111230] [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: 12/28/2023] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Despite decades of research, endometriosis remains a mysterious gynecological disease with unknown etiology and pathogenesis. Krüppel-like Factor 6 (KLF6), a transcription factor, has a wide expression profile and regulates a variety of biological processes. Here, we investigated the expression and function of KLF6 and its possible regulatory mechanisms in endometriosis. To determine the function of KLF6, knockdown and overexpression experiments were performed in eutopic endometrial stromal cells (EU-ESCs) and ectopic endometrial stromal cells (EC-ESCs), respectively. Cell viability, apoptosis, migration, invasion, and angiogenesis assays were conducted in ESCs. ChIP-sequencing and mRNA-sequencing were performed to investigate the functional mechanism of KLF6 in regulating ESCs. We found that KLF6 was highly expressed in eutopic endometrium of endometriosis patients, compared with ectopic endometrium. Similarly, the same was true in EU-ESCs, which was compared with EC-ESCs. Overexpression of KLF6 significantly suppressed EC-ESC proliferation, migration and invasion and induced cell apoptosis, while knockdown of KLF6 resulted in the opposite effects on EU-ESCs. Overexpression of KLF6 significantly inhibited EC-ESC angiogenesis. Mechanistically, the results of ChIP sequencing and mRNA sequencing revealed that CTNNB1 may be a transcriptional target regulated by KLF6. Reintroduction of KLF6 reversed the effects of KLF6 knockdown on EU-ESCs. KLF6 inhibited the proliferation, migration and angiogenesis of EC-ESCs by inhibiting the expression of CTNNB1. Our findings provided a new perspective on the role of KLF6 in endometriosis progression and inspire potential targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jingwen Shi
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Wenda Jing
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Yueyun He
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Ying Huang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
5
|
Brinca AT, Peiró AM, Evangelio PM, Eleno I, Oliani AH, Silva V, Vicente LF, Ramalhinho AC, Gallardo E. Follicular Fluid and Blood Monitorization of Infertility Biomarkers in Women with Endometriosis. Int J Mol Sci 2024; 25:7177. [PMID: 39000283 PMCID: PMC11241429 DOI: 10.3390/ijms25137177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Infertility is recognized globally as a social disease and a growing medical condition, posing a significant challenge to modern reproductive health. Endometriosis, the third-most frequent gynecologic disorder, is one of the most common and intricate conditions that can lead to female infertility. Despite extensive research, the etiology, malignant transformation, and biological therapy of endometriosis remain unknown. Blood and follicular fluid are two matrices that have been carefully studied and can provide insights into women's health. These matrices are clinically significant because they contain metabolites closely associated with women's illness stage and reproductive outcomes. Nowadays, the application of metabolomic analysis in biological matrices may be able to predict the outcome of assisted reproductive technologies with greater precision. From a molecular viewpoint on reproductive health, we evaluate and compare the utilization of human follicular fluid and blood as matrices in analysis for diagnostic and assisted reproductive technology (ART) predictors of success for endometriosis patients. In the follicular fluid (FF), plasma, and serum of endometriosis-affected women, researchers identified dysregulations of oxidative stress, upregulation of several immune factors, and aberrations in energy metabolic pathways. The altered signatures negatively correlate with the overall oocyte and embryo quality and fertilization rate.
Collapse
Affiliation(s)
- Ana Teresa Brinca
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal;
| | - Ana Maria Peiró
- Pharmacogenetic Unit, Clinical Pharmacology Department, Alicante Institute for Health and Biomedical Research (ISABIAL), Dr. Balmis General University Hospital, 03010 Alicante, Spain;
- Institute of Bioengineering, Miguel Hernández University, 03202 Elche, Spain
| | | | - Irene Eleno
- Unidad de Reproduccion, Servicio de Ginecologia y Obstetricia, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain;
| | - Antonio Helio Oliani
- Assisted Reproduction Laboratory, Cova da Beira Local Health Unit, 6200-251 Covilhã, Portugal;
- São José do Rio Preto School of Medicine, Gynaecology and Obstetrics, São José do Rio Preto 15090-000, Brazil
| | - Vladimiro Silva
- Ferticentro—Centro de Estudos de Fertilidade S.A., 3000-316 Coimbra, Portugal;
- Procriar—Centro de Procriação Medicamente Assistida, 4100-130 Porto, Portugal
| | | | - Ana Cristina Ramalhinho
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal;
- Assisted Reproduction Laboratory, Cova da Beira Local Health Unit, 6200-251 Covilhã, Portugal;
| | - Eugenia Gallardo
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal;
- Laboratório de Fármaco-Toxicologia, UBIMedical, University of Beira Interior, 6200-284 Covilhã, Portugal
| |
Collapse
|
6
|
Artemova D, Vishnyakova P, Gantsova E, Elchaninov A, Fatkhudinov T, Sukhikh G. The prospects of cell therapy for endometriosis. J Assist Reprod Genet 2023; 40:955-967. [PMID: 36964451 PMCID: PMC10239410 DOI: 10.1007/s10815-023-02772-5] [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] [Indexed: 03/26/2023] Open
Abstract
Endometriosis is a chronic inflammatory estrogen-dependent disease characterized by the growth of endometrial-like tissue outside the physiological region. Despite the fact that this disease is common, laparoscopic surgery is currently the gold standard in the treatment of endometriosis. In this regard, it is necessary to develop new effective methods of minimally invasive therapy for endometriosis. One of the promising areas in the treatment of endometriosis is cell therapy. Cellular therapy is a vast branch of therapeutic methods with various agents. Potential cell therapies for endometriosis may be based on the principle of targeting aspects of the pathogenesis of the disease: suppression of estrogen receptor activity, angiogenesis, fibrosis, and a decrease in the content of stem cells in endometriosis foci. In addition, immune cells such as NK cells and macrophages may be promising agents for cell therapy of endometriosis. Standing apart in the methods of cell therapy is the replacement therapy of endometriosis. Thus, many studies in the field of the pathogenesis of endometriosis can shed light not only on the causes of the disease and may contribute to the development of new methods for personalized cell therapy of endometriosis.
Collapse
Affiliation(s)
- Daria Artemova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - Polina Vishnyakova
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Elena Gantsova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - Andrey Elchaninov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia.
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia.
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| |
Collapse
|
7
|
Ning WJ, Liu X, Zeng HY, An ZQ, Luo WX, Xia NS. Recent progress in antibody-based therapeutics for triple-negative breast cancer. Expert Opin Drug Deliv 2022; 19:815-832. [PMID: 35738312 DOI: 10.1080/17425247.2022.2093853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is a subtype of severely aggressive breast cancer that lacks the expression of oestrogen receptor (ER), progesterone receptor and human epidermal growth factor receptor 2 (HER2) and is highly metastatic and related to a poor prognosis. Current standard treatments are still limited to systemic chemotherapy, radiotherapy, and surgical resection. More effective treatments are urgently needed. AREAS COVERED The immunogenicity of TNBC has provided opportunities for the development of targeted immunotherapy. In this review, we focus on the recent development in antibody-based drug modalities, including angiogenesis inhibitors, immune checkpoint inhibitors, antibody-drug conjugates, immunoconjugates, T cell-redirecting bispecific antibodies and CAR-T cells, and their mechanisms of action in TNBC. EXPERT OPINION At present, the treatment of TNBC is still a major challenge that needs to be addressed. Novel immunotherapies are promising opportunities for improving the management of this aggressive disease.
Collapse
Affiliation(s)
- Wen-Jing Ning
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
| | - Xue Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
| | - Hong-Ye Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
| | - Zhi-Qiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wen-Xin Luo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
| | - Ning-Shao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
8
|
Chung MS, Han SJ. Endometriosis-Associated Angiogenesis and Anti-angiogenic Therapy for Endometriosis. Front Glob Womens Health 2022; 3:856316. [PMID: 35449709 PMCID: PMC9016174 DOI: 10.3389/fgwh.2022.856316] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/24/2022] [Indexed: 01/02/2023] Open
Abstract
Endometriosis is a known estrogen-dependent inflammatory disease affecting reproductive-aged women. Common symptoms include pelvic pain, dysmenorrhea, dyspareunia, heavy menstrual bleeding, and infertility. The exact etiology of endometriosis is largely unknown, and, thus, the diagnosis and treatment of endometriosis are challenging. A complex interplay of many molecular mechanisms is thought to aid in the progression of endometriosis, most notably angiogenesis. This mini-review examines our current knowledge of the molecular etiology of endometriosis-associated angiogenesis and discusses anti-angiogenic therapy, in the blockade of endometriosis-associated angiogenesis, as potential non-hormonal therapy for the treatment of endometriosis.
Collapse
Affiliation(s)
- Monica S. Chung
- Division of Reproductive Endocrinology and Infertility, Department of Ob/Gyn, Baylor College of Medicine, Houston, TX, United States
| | - Sang Jun Han
- Laboratory of Dan L. Duncan Cancer Center and Reproductive Medicine, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Sang Jun Han
| |
Collapse
|
9
|
Załęcka J, Pankiewicz K, Issat T, Laudański P. Molecular Mechanisms Underlying the Association between Endometriosis and Ectopic Pregnancy. Int J Mol Sci 2022; 23:ijms23073490. [PMID: 35408850 PMCID: PMC8998627 DOI: 10.3390/ijms23073490] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
Endometriosis is a common inflammatory disease characterized by the presence of endometrial cells outside the uterine cavity. It is estimated that it affects 10% of women of reproductive age. Its pathogenesis covers a wide range of abnormalities, including adhesion, proliferation, and cell signaling disturbances. It is associated with a significant deterioration in quality of life as a result of chronic pelvic pain and may also lead to infertility. One of the most serious complications of endometriosis is an ectopic pregnancy (EP). Currently, the exact mechanism explaining this phenomenon is unknown; therefore, there are no effective methods of prevention. It is assumed that the pathogenesis of EP is influenced by abnormalities in the contraction of the fallopian tube muscles, the mobility of the cilia, and in the fallopian microenvironment. Endometriosis can disrupt function on all three levels and thus contribute to the implantation of the embryo beyond the physiological site. This review takes into account aspects of the molecular mechanisms involved in the pathophysiology of endometriosis and EP, with particular emphasis on the similarities between them.
Collapse
Affiliation(s)
- Julia Załęcka
- 1st Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland;
| | - Katarzyna Pankiewicz
- Department of Obstetrics and Gynecology, Institute of Mother and Child in Warsaw, Kasprzaka 17a, 01-211 Warsaw, Poland; (K.P.); (T.I.)
| | - Tadeusz Issat
- Department of Obstetrics and Gynecology, Institute of Mother and Child in Warsaw, Kasprzaka 17a, 01-211 Warsaw, Poland; (K.P.); (T.I.)
| | - Piotr Laudański
- 1st Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland;
- OVIklinika Infertility Center, Połczyńska 31, 01-377 Warsaw, Poland
- Correspondence:
| |
Collapse
|
10
|
Wang X, Wu P, Zeng C, Zhu J, Zhou Y, Lu Y, Xue Q. Long Intergenic Non-Protein Coding RNA 02381 Promotes the Proliferation and Invasion of Ovarian Endometrial Stromal Cells through the miR-27b-3p/CTNNB1 Axis. Genes (Basel) 2022; 13:433. [PMID: 35327987 PMCID: PMC8955621 DOI: 10.3390/genes13030433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/20/2022] [Accepted: 02/24/2022] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Catenin Beta 1 (CTNNB1) is a key regulator of cell proliferation and invasion in endometriosis; however, its upstream factor is not clear. Long noncoding RNAs may participate in endometriosis. The aim of this study was to investigate the mechanism of interaction between LINC02381 and CTNNB1 in endometriosis. METHOD Screening and validation of RNAs were completed by whole transcriptional sequencing and qRT-PCR. The subcellular localization of LINC02381 was determined by RNA in situ hybridization and nucleo-cytoplasmic separation. Plasmids were transfected for functional experiments. Luciferase assay was used to verify the binding relationship. RESULTS The expression of LINC02381 and CTNNB1 was significantly increased in ovarian ectopic endometrial tissues (OSAs) and ectopic endometrial stromal cells (ESCs). When LINC02381 was downregulated in ESCs, the expression of CTNNB1, metallopeptidase 9 (MMP9) and cyclinD1, as well as ESCs invasion and proliferation, decreased. LINC02381 was mainly present in the cytoplasm of ESCs, indicating that it may act as a competitive endogenous RNA. Bioinformatic analysis revealed that microRNA-27b-3p (miR-27b-3p) is a downstream target of LINC02381. miR-27b-3p decreased in OSAs and ESCs. Moreover, when miR-27b-3p was upregulated in ESCs, the expression of CTNNB1, MMP9 and cyclinD1, as well as the invasion and proliferation ability of ESCs, were reduced. Additionally, rescue experiments demonstrated that the expression of CTNNB1, MMP9 and cyclinD1, as well as the invasion and proliferation ability, were significantly increased in the group transfected with both sh-LINC02381 and a miR-27b-3p inhibitor. CONCLUSION LINC02381 upregulated CTNNB1 by adsorbing miR-27b-3p, causing increased proliferation and invasion of ESCs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qing Xue
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China; (X.W.); (P.W.); (C.Z.); (J.Z.); (Y.Z.); (Y.L.)
| |
Collapse
|
11
|
Eades W, Liu W, Shen Y, Shi Z, Yan B. Covalent CES2 Inhibitors Protect against Reduced Formation of Intestinal Organoids by the Anticancer Drug Irinotecan. Curr Drug Metab 2022; 23:1000-1010. [PMID: 36515038 PMCID: PMC10258227 DOI: 10.2174/1389200224666221212143904] [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: 07/21/2022] [Revised: 10/26/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Irinotecan is widely used to treat various types of solid and metastatic cancer. It is an ester prodrug and its hydrolytic metabolite (SN-38) exerts potent anticancer activity. Irinotecan is hydrolyzed primarily by carboxylesterase-2 (CES2), a hydrolase abundantly present in the intestine such as the duodenum. We have identified several potent and covalent CES2 inhibitors such as remdesivir and sofosbuvir. Remdesivir is the first small molecule drug approved for COVID-19, whereas sofosbuvir is a paradigm-shift medicine for hepatitis C viral infection. Irinotecan is generally well-tolerated but associated with severe/life-threatening diarrhea due to intestinal accumulation of SN-38. OBJECTIVE This study was to test the hypothesis that remdesivir and sofosbuvir protect against irinotecan-induced epithelial injury associated with gastrointestinal toxicity. METHODS To test this hypothesis, formation of organoids derived from mouse duodenal crypts, a robust cellular model for intestinal regeneration, was induced in the presence or absence of irinotecan +/- pretreatment with a CES2 drug inhibitor. RESULTS Irinotecan profoundly inhibited the formation of intestinal organoids and the magnitude of the inhibition was greater with female crypts than their male counterparts. Consistently, crypts from female mice had significantly higher hydrolytic activity toward irinotecan. Critically, remdesivir and sofosbuvir both reduced irinotecan hydrolysis and reversed irinotecan-reduced formation of organoids. Human duodenal samples robustly hydrolyzed irinotecan, stable CES2 transfection induced cytotoxicity and the cytotoxicity was reduced by CES2 drug inhibitor. CONCLUSION These findings establish a therapeutic rationale to reduce irinotecan-gastrointestinal injury and serve as a cellular foundation to develop oral formulations of irinotecan with high safety.
Collapse
Affiliation(s)
- William Eades
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
- Equal contribution
| | - William Liu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
- Equal contribution
| | - Yue Shen
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
- Equal contribution
| | - Zhanquan Shi
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
12
|
Estrogens Regulate Placental Angiogenesis in Horses. Int J Mol Sci 2021; 22:ijms222212116. [PMID: 34829994 PMCID: PMC8621320 DOI: 10.3390/ijms222212116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
A sufficient vascular network within the feto-maternal interface is necessary for placental function. Several pregnancy abnormalities have been associated with abnormal vascular formations in the placenta. We hypothesized that growth and expansion of the placental vascular network in the equine (Equus caballus) placenta is regulated by estrogens (estrogen family hormones), a hormone with a high circulating concentration during equine gestation. Administration of letrozole, a potent and specific inhibitor of aromatase, during the first trimester (D30 to D118), decreased circulatory estrone sulfate concentrations, increased circulatory testosterone and androstenedione concentrations, and tended to reduce the weight of the fetus (p < 0.1). Moreover, the gene expression of CYP17A1 was increased, and the expression of androgen receptor was decreased in the D120 chorioallantois (CA) of letrozole-treated mares in comparison to that of the control mares. We also found that at D120, the number of vessels tended to decrease in the CAs with letrozole treatment (p = 0.07). In addition, expression of a subset of angiogenic genes, such as ANGPT1, VEGF, and NOS2, were altered in the CAs of letrozole-treated mares. We further demonstrated that 17β-estradiol increases the expression of ANGPT1 and VEGF and increases the angiogenic activity of equine endothelial cells in vitro. Our results from the estrogen-suppressed group demonstrated an impaired placental vascular network, suggesting an estrogen-dependent vasculogenesis in the equine CA during the first trimester.
Collapse
|
13
|
Giacomini E, Minetto S, Li Piani L, Pagliardini L, Somigliana E, Viganò P. Genetics and Inflammation in Endometriosis: Improving Knowledge for Development of New Pharmacological Strategies. Int J Mol Sci 2021; 22:ijms22169033. [PMID: 34445738 PMCID: PMC8396487 DOI: 10.3390/ijms22169033] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
According to a rich body of literature, immune cell dysfunctions, both locally and systemically, and an inflammatory environment characterize all forms of endometriosis. Alterations in transcripts and proteins involved in the recruitment of immune cells, in the interaction between cytokines and their receptors, cellular adhesion and apoptosis have been demonstrated in endometriotic lesions. The objective of this narrative review is to provide an overview of the components and mechanisms at the intersection between inflammation and genetics that may constitute vanguard therapeutic approaches in endometriosis. The GWAS technology and pathway-based analysis highlighted the role of the MAPK and the WNT/β-catenin cascades in the pathogenesis of endometriosis. These signaling pathways have been suggested to interfere with the disease establishment via several mechanisms, including apoptosis, migration and angiogenesis. Extracellular vesicle-associated molecules may be not only interesting to explain some aspects of endometriosis progression, but they may also serve as therapeutic regimens per se. Immune/inflammatory dysfunctions have always represented attractive therapeutic targets in endometriosis. These would be even more interesting if genetic evidence supported the involvement of functional pathways at the basis of these alterations. Targeting these dysfunctions through next-generation inhibitors can constitute a therapeutic alternative for endometriosis.
Collapse
Affiliation(s)
- Elisa Giacomini
- Reproductive Sciences Laboratory, Obstetrics and Gynecology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (E.G.); (L.P.)
| | - Sabrina Minetto
- Obstetrics and Gynecology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Letizia Li Piani
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (L.L.P.); (E.S.)
| | - Luca Pagliardini
- Reproductive Sciences Laboratory, Obstetrics and Gynecology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (E.G.); (L.P.)
| | - Edgardo Somigliana
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (L.L.P.); (E.S.)
| | - Paola Viganò
- Infertility Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-5503-4302
| |
Collapse
|
14
|
Alonso-Diez Á, Cáceres S, Peña L, Crespo B, Illera JC. Anti-Angiogenic Treatments Interact with Steroid Secretion in Inflammatory Breast Cancer Triple Negative Cell Lines. Cancers (Basel) 2021; 13:3668. [PMID: 34359570 PMCID: PMC8345132 DOI: 10.3390/cancers13153668] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/21/2022] Open
Abstract
Human inflammatory breast cancer (IBC) is a highly angiogenic disease for which antiangiogenic therapy has demonstrated only a modest response, and the reason for this remains unknown. Thus, the purpose of this study was to determine the influence of different antiangiogenic therapies on in vitro and in vivo steroid hormone and angiogenic growth factor production using canine and human inflammatory breast carcinoma cell lines as well as the possible involvement of sex steroid hormones in angiogenesis. IPC-366 and SUM149 cell lines and xenotransplanted mice were treated with different concentrations of VEGF, SU5416, bevacizumab and celecoxib. Steroid hormone (progesterone, dehydroepiandrostenedione, androstenedione, testosterone, dihydrotestosterone, estrone sulphate and 17β-oestradiol), angiogenic growth factors (VEGF-A, VEGF-C and VEGF-D) and IL-8 determinations in culture media, tumour homogenate and serum samples were assayed by EIA. In vitro, progesterone- and 17β-oestradiol-induced VEGF production promoting cell proliferation and androgens are involved in the formation of vascular-like structures. In vivo, intratumoural testosterone concentrations were augmented and possibly associated with decreased metastatic rates, whereas elevated E1SO4 concentrations could promote tumour progression after antiangiogenic therapies. In conclusion, sex steroid hormones could regulate the production of angiogenic factors. The intratumoural measurement of sex steroids and growth factors may be useful to develop preventive and individualized therapeutic strategies.
Collapse
Affiliation(s)
- Ángela Alonso-Diez
- Department Animal Medicine, Surgery and Pathology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Sara Cáceres
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Laura Peña
- Department Animal Medicine, Surgery and Pathology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Belén Crespo
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Juan Carlos Illera
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| |
Collapse
|
15
|
Lin TC, Wang KH, Chuang KH, Kao AP, Kuo TC. Interleukin-33 promotes invasiveness of human ovarian endometriotic stromal cells through the ST2/MAPK/MMP-9 pathway activated by 17β-estradiol. Taiwan J Obstet Gynecol 2021; 60:658-664. [PMID: 34247803 DOI: 10.1016/j.tjog.2021.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVE Endometriosis is an estrogen-dependent, benign, and chronic gynecological disorder occurring in women of reproductive age. Although the pathogenesis of endometriosis is poorly understood, implantation theory indicates that viable endometrial cells shed from the endometrium into the pelvic peritoneum or ovaries, possibly through retrograde menstruation, and then reattach, invade, and damage other tissues. Interleukin (IL)-33, a new member of the IL-1 superfamily, is mainly upregulated by stromal cells following proinflammatory stimulation. Matrix metalloproteinases (MMPs) are involved in the degradation and reconstruction of the extracellular matrix. MMP-9 participates in the pathogenesis of endometriosis by promoting the invasion of endometriotic cells. This study investigated the effect of IL-33 on the cell invasion ability of and MMP-9 expression in human stromal cells derived from ovarian endometrioma (hOVEN-SCs). MATERIALS AND METHODS We isolated hOVEN-SCs from human ovarian endometrioma. Gene expression was analyzed using the Illumina Human WG-6 v2 Expression BeadChips microarray platform and through reverse transcription-polymerase chain reaction. Cell migration and invasion were examined by performing the transwell chamber assay. RESULTS We found that 17β-estradiol could increase the expression of IL-33 and ST2 through the estrogen receptor pathway in hOVEN-SCs. Moreover, IL-33 upregulated MMP-9 expression in and enhanced the invasion ability of hOVEN-SCs through the ST2/MAPK signaling pathway. Our results showed that MMP-9 expression was essential for IL-33-induced cell invasion. CONCLUSION Our main finding is that 17β-estradiol could increase IL-33 expression through the estrogen receptor pathway and activate MMP-9 expression in and invasion ability of hOVEN-SCs through the IL-33/ST2/MAPK signaling pathway. The results of this study and further related studies may provide new strategies for the prevention and treatment of endometriosis.
Collapse
Affiliation(s)
- Ta-Chin Lin
- Department of Obstetrics and Gynecology, Kuo General Hospital, Tainan, Taiwan; Center for Reproductive Medicine, Kuo General Hospital, Tainan, Taiwan
| | - Kai-Hung Wang
- Department of Obstetrics and Gynecology, Kuo General Hospital, Tainan, Taiwan; Center for Reproductive Medicine, Kuo General Hospital, Tainan, Taiwan; Department of Laboratory Medicine, Kuo General Hospital, Tainan, Taiwan.
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - An-Pei Kao
- Stemforce Biotechnology Co., Ltd, Chiayi, Taiwan
| | - Tsung-Cheng Kuo
- Department of Obstetrics and Gynecology, Kuo General Hospital, Tainan, Taiwan; Center for Reproductive Medicine, Kuo General Hospital, Tainan, Taiwan
| |
Collapse
|
16
|
El Sabeh M, Saha SK, Afrin S, Islam MS, Borahay MA. Wnt/β-catenin signaling pathway in uterine leiomyoma: role in tumor biology and targeting opportunities. Mol Cell Biochem 2021; 476:3513-3536. [PMID: 33999334 DOI: 10.1007/s11010-021-04174-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
Uterine leiomyoma is the most common tumor of the female reproductive system and originates from a single transformed myometrial smooth muscle cell. Despite the immense medical, psychosocial, and financial impact, the exact underlying mechanisms of leiomyoma pathobiology are poorly understood. Alterations of signaling pathways are thought to be instrumental in leiomyoma biology. Wnt/β-catenin pathway appears to be involved in several aspects of the genesis of leiomyomas. For example, Wnt5b is overexpressed in leiomyoma, and the Wnt/β-catenin pathway appears to mediate the role of MED12 mutations, the most common mutations in leiomyoma, in tumorigenesis. Moreover, Wnt/β-catenin pathway plays a paracrine role where estrogen/progesterone treatment of mature myometrial or leiomyoma cells leads to increased expression of Wnt11 and Wnt16, which induces proliferation of leiomyoma stem cells and tumor growth. Constitutive activation of β-catenin leads to myometrial hyperplasia and leiomyoma-like lesions in animal models. Wnt/β-catenin signaling is also closely involved in mechanotransduction and extracellular matrix regulation and relevant alterations in leiomyoma, and crosstalk is noted between Wnt/β-catenin signaling and other pathways known to regulate leiomyoma development and growth such as estrogen, progesterone, TGFβ, PI3K/Akt/mTOR, Ras/Raf/MEK/ERK, IGF, Hippo, and Notch signaling. Finally, evidence suggests that inhibition of the canonical Wnt pathway using β-catenin inhibitors inhibits leiomyoma cell proliferation. Understanding the molecular mechanisms of leiomyoma development is essential for effective treatment. The specific Wnt/β-catenin pathway molecules discussed in this review constitute compelling candidates for therapeutic targeting.
Collapse
Affiliation(s)
- Malak El Sabeh
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Subbroto Kumar Saha
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Sadia Afrin
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Md Soriful Islam
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Mostafa A Borahay
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
| |
Collapse
|
17
|
Role of Vascular Endothelial Growth Factor (VEGF) in Human Embryo Implantation: Clinical Implications. Biomolecules 2021; 11:biom11020253. [PMID: 33578823 PMCID: PMC7916576 DOI: 10.3390/biom11020253] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is a well-known angiogenic factor that plays a critical role in various physiological and pathological processes. VEGF also contributes to the process of embryo implantation by enhancing embryo development, improving endometrial receptivity, and facilitating the interactions between the developing embryo and the endometrium. There is a correlation between the alteration of VEGF expression and reproductive failure, including recurrent implantation failure (RIF) and recurrent miscarriage (RM). In order to clarify the role of VEGF in embryo implantation, we reviewed recent literature concerning the expression and function of VEGF in the reproductive system around the time of embryo implantation and we provide a summary of the findings reported so far. We also explored the effects and the possible underlying mechanisms of action of VEGF in embryo implantation.
Collapse
|
18
|
Liang X, Su Y, Huo Y. Forkhead box protein O1 (FoxO1) /SERPINB1 ameliorates ROS production in diabetic nephropathy. Food Sci Nutr 2021; 9:44-51. [PMID: 33473269 PMCID: PMC7802531 DOI: 10.1002/fsn3.1859] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 11/30/2022] Open
Abstract
With the increasing prevalence of diabetes in recent years, diabetic nephropathy (DN) has become a severe disease that greatly threatens human health. DN not only is a common complication of diabetes, but also takes an important place in kidney disease. To this end, the present study was designed to explore the effects of Forkhead box protein O1 (FoxO1) on reactive oxygen species (ROS) production in DN mice. DN mice were treated with recombinant protein of FoxO1. Afterward, inflammation ELISA kits were used to measure the levels of TNF-α, IL-1β, IL-6, and IL-18. The levels of MDA, SOD, GSH, and GSH-PX were measured using kits according to the manufacturer's instructions. In addition, the production of ROS was assessed. Interestingly, the expression of FoxO1 was down-regulated in DN mice. The treatment of FoxO1 recombinant protein ameliorated MDA levels, increased the levels of SOD, GSH, and GSH-PX, and induced both mRNA and protein expression of hepatic serine protease inhibitor B1 (serpinB1) in ND mice. Similarly, FoxO1 reduced MDA levels and ROS production, increased the levels of SOD, GSH, and GSH-PXs, and induced the mRNA and protein expression of serpinB1 in in vitro model of DN. The inhibition of serpinB1 attenuated the effects of FoxO1 on ROS production-induced oxidative stress in in vitro model of DN. Overall, FoxO1/SERPINB1 ameliorated ROS production-induced oxidative stress in DN.
Collapse
Affiliation(s)
- Xiaoya Liang
- Infection management departmentAffiliated hospital of shaanxi university of traditional Chinese medicineXianyangShaanxiChina
| | - Yanjin Su
- The first Department of EndocrinologyShaanxi university of traditional Chinese medicineXianyangShaanxiChina
| | - Yongbo Huo
- Internal Medicine DepartmentYan'an Hospital of traditional Chinese MedicineShaanxiShaanxiChina
| |
Collapse
|
19
|
Orazov M, Abitova M, Khamoshina M, Volkova S, Aryutin D, Shustova V. OVARIAN ENDOMETRIOSIS: THE MODERN POSSIBILITIES OF RELAPSE PREDICTION. REPRODUCTIVE MEDICINE 2020. [DOI: 10.37800/rm2020-1-30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The article summarizes the current understanding of risk factors and mechanisms of ovarian endometriosis relapses,
This article summarizes the current understanding of risk factors for relapse, mechanisms of recurrence of endometrioid ovarian cysts, as well as the potential for their prediction using genetic and molecular-biological predictors.
Collapse
|
20
|
He X, Liu N, Mu T, Lu D, Jia C, Wang S, Yin C, Liu L, Zhou L, Huang X, Ma Y. Oestrogen induces epithelial-mesenchymal transition in endometriosis via circ_0004712/miR-148a-3p sponge function. J Cell Mol Med 2020; 24:9658-9666. [PMID: 32667746 PMCID: PMC7520264 DOI: 10.1111/jcmm.15495] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/14/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Endometriosis is a common, chronic gynaecologic disease affecting up to 10% of women in their reproductive age and leading to pain and infertility. Oestrogen (E2)‐induced epithelial‐mesenchymal transition (EMT) process has been considered as a key factor of endometriosis development. Recently, the dysregulated circular RNAs (circRNAs) have been discovered in endometriosis tissues. However, the molecular mechanism of circRNAs on the E2‐induced EMT process in endometriosis is still unknown. Here, we demonstrated that circ_0004712 up‐regulated by E2 treatment in endometrial epithelial cells. Knock‐down the expression of circ_0004712 significantly suppressed E2‐induced cell migration activity. Meanwhile, we identified miR‐148a‐3p as a potential target miRNA of circ_0004712. Inhibited the expression of miR‐148a‐3p could recovered the effect of circ_0004712 knock‐down in E2‐treated endometrial epithelial. Furthermore, Western blot assay showed that E2 treatment could increase the expression and activity of β‐catenin, snail and N‐cadherin and reduce the expression of E‐cadherin. The expression and activity of β‐catenin pathway were recovered by circ_0004712 knock‐down or miR‐148a‐3p overexpression. Altogether, the results demonstrate that circ_0004712/miR‐148a‐3p plays an important role in E2‐induced EMT process in the development of endometriosis, and the molecular mechanism may be associated with the β‐catenin pathway. This work highlighted the importance of circRNAs in the development of endometriosis and provide a new biomarker for diagnosis and therapies.
Collapse
Affiliation(s)
- Xin He
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Nana Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Tianyi Mu
- Department of Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Dan Lu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Chanwei Jia
- Department of Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Shuyu Wang
- Department of Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Chenghong Yin
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Lingyan Liu
- College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Liying Zhou
- Department of Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Xiaowu Huang
- Department of Hysteroscopic Center, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Yanmin Ma
- Department of Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Dihm K, Ek M, Roth B, Ohlsson B. Plasma AXIN1 expression exhibit negative correlations with inflammatory biomarkers and is associated with gastrointestinal symptoms in endometriosis. Biomed Rep 2020; 12:211-221. [PMID: 32257184 PMCID: PMC7100128 DOI: 10.3892/br.2020.1282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
The cytoplasmic protein AXIN1 is involved in the Wnt signalling pathway and its expression is increased in patients with endometriosis compared with healthy controls. The aim of the present cross-sectional study was to further assess the levels of AXIN1 and other inflammatory biomarkers in patients with endometriosis. Patients with laparoscopy-verified endometriosis were recruited (n=172) and completed a questionnaire regarding socioeconomic factors, lifestyle habits and medical history. Plasma AXIN1 and high-sensitivity C-reactive protein (hs-CRP) levels were analysed by ELISA. The levels of calprotectin were determined in the faeces, and the haemoglobin concentration and number of erythrocytes, leukocytes and platelets were determined in the blood in a subgroup of 64 patients during clinical routine procedures. F-calprotectin expression was detected in 18 women (28.1%), who had more severe constipation and more frequently experienced incomplete evacuation when defecating, and 5 women (7.8%) exhibited elevated levels. P-AXIN1 levels were higher in patients who received hormonal treatment, and correlated inversely with faecal-calprotectin levels (P=0.003), B-haemoglobin levels (P=0.030) and the numbers of B-erythrocytes (P=0.033) and B-platelets (P=0.017), but were not correlated with hs-CRP levels (P=0.818). Higher levels of AXIN1 were associated with the duration of the gastrointestinal symptoms and with diarrhoea, constipation, vomiting and nausea and the intestinal symptoms' effect on quality of life, and tended to be associated with the duration of endometriosis. Hs-CRP expression was not associated with the clinical characteristics or symptoms of endometriosis, but higher levels were associated with obesity (P=0.002) and hormonal treatment (P=0.011). In conclusion, P-AXIN1 expression was negatively correlated with certain inflammatory biomarkers and was positively associated with gastrointestinal symptoms. P-AXIN1 levels were increased in patients who received hormonal treatment, highlighting the importance of obtaining native samples for future studies regarding its role in the development and presentation of endometriosis. However, hs-CRP and other studied biomarkers seemed to be of no value for the assessment and diagnosis of endometriosis.
Collapse
Affiliation(s)
- Katharina Dihm
- Department of Internal Medicine, Skane University Hospital, Lund University, 205 02 Malmo, Sweden
| | - Malin Ek
- Department of Internal Medicine, Skane University Hospital, Lund University, 205 02 Malmo, Sweden
| | - Bodil Roth
- Department of Internal Medicine, Skane University Hospital, Lund University, 205 02 Malmo, Sweden
| | - Bodil Ohlsson
- Department of Internal Medicine, Skane University Hospital, Lund University, 205 02 Malmo, Sweden
| |
Collapse
|
22
|
Ali M, Shahin SM, Sabri NA, Al-Hendy A, Yang Q. Activation of β-Catenin Signaling and its Crosstalk With Estrogen and Histone Deacetylases in Human Uterine Fibroids. J Clin Endocrinol Metab 2020; 105:5639769. [PMID: 31761932 PMCID: PMC7064306 DOI: 10.1210/clinem/dgz227] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/23/2019] [Indexed: 12/23/2022]
Abstract
CONTEXT Uterine fibroids (UF) are the most common benign tumor of the myometrium (MM) in women of reproductive age. However, the mechanism underlying the pathogenesis of UF is largely unknown. OBJECTIVE To explore the link between nuclear β-catenin and UF phenotype and β-catenin crosstalk with estrogen and histone deacetylases (HDACs). DESIGN Protein/RNA levels of β-catenin (CTNNB1 gene), its responsive markers cyclin D1 and c-Myc, androgen receptor (AR), p27, and class-I HDACs were measured in matched UF/MM tissues or cell populations. The effects of chemical inhibition/activation and genetic knockdown of CTNNB1 on UF phenotype were measured. The anti-UF effect of 2 HDAC inhibitors was evaluated. MAIN OUTCOME MEASURE β-catenin nuclear translocation in response to β-catenin inhibition/activation, estrogen, and HDAC inhibitors in UF cells. RESULTS UF tissues/cells showed significantly higher expression of nuclear β-catenin, cyclin D1, c-Myc, and HDACs 1, 2, 3, and 8 than MM. Estradiol induced β-catenin nuclear translocation and consequently its responsive genes in both MM and UF cells, while an estrogen receptor antagonist reversed this induction effect. Treatment with β-catenin or HDAC inhibitors led to dose-dependent growth inhibition, while Wnt3a treatment increased proliferation compared with control. Chemical inhibition of β-catenin decreased cyclin D1 and c-Myc expression levels, while β-catenin activation increased expression of the same markers. Genetic knockdown of CTNNB1 resulted in a marked decrease in β-catenin, cyclin D1, c-Myc, and AR expression. Treatment of UF cells with HDAC inhibitors decreased nuclear β-catenin, cyclin D1, and c-Myc expression. Moreover, HDAC inhibitors induced apoptosis of UF cells and cell cycle arrest. CONCLUSION β-catenin nuclear translocation contributes to UF phenotype, and β-catenin signaling is modulated by estradiol and HDAC activity.
Collapse
Affiliation(s)
- Mohamed Ali
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois
- Clinical Pharmacy Department, Faculty of Pharmacy, ASU, Cairo, Egypt
| | | | - Nagwa Ali Sabri
- Clinical Pharmacy Department, Faculty of Pharmacy, ASU, Cairo, Egypt
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois
- Correspondence: Dr Qiwei Yang, 1Department of Obstetrics and Gynecology, University of Illinois at Chicago, 909 S. Wolcott Ave, Chicago, IL 60612, USA. E-mail:
| |
Collapse
|
23
|
Weng LC, Hou SH, Lei ST, Peng HY, Li MQ, Zhao D. Estrogen-regulated CD200 inhibits macrophage phagocytosis in endometriosis. J Reprod Immunol 2020; 138:103090. [PMID: 32014721 DOI: 10.1016/j.jri.2020.103090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Endometriosis (EMS) is a benign disease that is related to estrogen, immune disorders and inflammation. The purpose of this research was to determine the expression of CD200 in EMS and to clarify its role in the pathogenesis of the disease. METHODS The levels of serum CD200 in patients with and without EMS were determined by ELISA. Furthermore, the expression of CD200 in normal eutopic endometrium and ectopic endometrium was detected by immunohistochemistry and western blotting. The CD200 receptor (CD200R) in macrophages in peritoneal fluid (pMØ) obtained from controls and patients with EMS was examined by western blotting. CD200 expression in human endometrial stromal cells (HESCs) stimulated with 17β-estradiol (E2) was measured by western blotting. Furthermore, macrophages were stimulated with different concentrations of CD200 and the effect on phagocytosis was analyzed. RESULTS The plasma CD200 levels of patients with EMS was significantly increased compared with controls (P = 0.0173, 95%CI [18.75, 159.6]). Compared with normal eutopic endometrium, the expression of CD200 was significantly increased in ectopic endometrial tissues. The CD200R expression in pMØ obtained from patients with EMS was increased compared with the controls (P = 0.0244). CD200 expression in HESCs stimulated with E2 was up-regulated. As the levels of CD200 increased, macrophage phagocytosis in vitro gradually decreased. CONCLUSIONS CD200 is an estrogen-induced molecule that impairs macrophage phagocytosis and may contribute to the immune escape of ectopic lesions in EMS.
Collapse
Affiliation(s)
- Li-Chun Weng
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| | - Shu-Hui Hou
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| | - Sha-Ting Lei
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| | - Hai-Yan Peng
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, No.1326, Pingliang Road, Shanghai, 200080, People's Republic of China.
| | - Dong Zhao
- Department of Obstetrics and Gynecology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Rd., Shanghai, 200011, People's Republic of China.
| |
Collapse
|
24
|
β-catenin signaling inhibitors ICG-001 and C-82 improve fibrosis in preclinical models of endometriosis. Sci Rep 2019; 9:20056. [PMID: 31882904 PMCID: PMC6934788 DOI: 10.1038/s41598-019-56302-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/30/2019] [Indexed: 12/13/2022] Open
Abstract
Endometriosis exhibits unique characteristics, such as fibrosis, resistance to apoptosis, and promotion of cell proliferation; however, its pathophysiology is not fully understood. Recurrence rates after treatment are high, and the progression risk continues until menopause; hence, more effective therapy for endometriosis is needed. CREB-binding protein (CBP)/β-catenin signaling inhibitors have demonstrated antifibrogenetic effects in liver, lung, and skin diseases. The present study evaluated the effects of two CBP/β-catenin signaling inhibitors, ICG-001 and C-82, on the progression of endometriosis using endometriotic cyst stromal cells from the ovary and normal endometrial stromal cells from the uterus. ICG-001 was also evaluated in a mouse model. ICG-001 and C-82 inhibited cell proliferation, fibrogenesis, and cell migration, and promoted apoptosis in vitro. ICG-001 inhibited the growth of endometriotic lesions in the mouse model. CBP/β-catenin signaling plays an important role in the pathophysiology of endometriosis. Inhibiting the CBP/β-catenin signal can be a therapeutic target for endometriosis.
Collapse
|
25
|
Shu C, Shu Y, Gao Y, Chi H, Han J. Inhibitory effect of AQP1 silencing on adhesion and angiogenesis in ectopic endometrial cells of mice with endometriosis through activating the Wnt signaling pathway. Cell Cycle 2019; 18:2026-2039. [PMID: 31251110 DOI: 10.1080/15384101.2019.1637202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The development mechanism of endometriosis remains unknown. Water channel aquaporin-1 (AQP1) enhances water flux across cell membranes, which is highly expressed and associated with cell migration, metastasis and angiogenesis in some human cancers. In this study, the role of the Wnt signaling pathway mediated by AQP1 in endometriosis was investigated, in a bid to provide new therapeutic targets for endometriosis. Microarray expression profiles were screened to acquired differentially expressed genes related to endometriosis. Mouse models with endometriosis were established and grouped. The level of endometriosis was evaluated by measurement of the volume of ectopic region. The expression of AQP1, pathway-related factors (Wnt1 and Wnt4), adhesion molecules (VCAM-1 and ICAM-1), invasive factors (MMP-2, MMP-9, TIMP-1 and TIMP-2), angiogenic factors (VEGF-A, VEGFR1 and VEGFR2) and apoptotic factors (Caspase-3, Caspase-9, Bax and BcL-2) was measured by RT-qPCR and western blot analysis. Furthermore, the role of AQP1 in adhesion, invasion, angiogenesis, and apoptosis of ectopic endometrial cells was determined by transfection of si-AQP1 plasmid. AQP1 was robustly expressed in endometriosis. AQP1 gene silencing alleviated the progression of endometriosis by activating the Wnt signaling pathway in mice with endometriosis. Specifically, silencing of AQP1 gene inhibited ectopic endometrial cell adhesion and invasion abilities, suppressed angiogenesis while promoted apoptosis. Collectively, the present study highlights the role of AQP1 in the regulation of the Wnt signaling pathway in endometriosis mouse models, suggesting that AQP1 could represent a new target aimed at improving the survival of patients with endometriosis.
Collapse
Affiliation(s)
- Chang Shu
- a Department of Obstetrics and Gynecology, The First Hospital of Jilin University , Changchun , P.R. China
| | - Yang Shu
- a Department of Obstetrics and Gynecology, The First Hospital of Jilin University , Changchun , P.R. China
| | - Yongmei Gao
- a Department of Obstetrics and Gynecology, The First Hospital of Jilin University , Changchun , P.R. China
| | - Hui Chi
- a Department of Obstetrics and Gynecology, The First Hospital of Jilin University , Changchun , P.R. China
| | - Jun Han
- b Department of Neonatology, The First Hospital of Jilin University , Changchun , P.R. China
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Postmenopausal endometriosis is a gynecologic disease, affecting 2-5% of postmenopausal woman. Current literature assessing the prevalence, pathogenesis, and treatment of this uncommon condition is limited, stressing the necessity for future research. This review examines the current literature on postmenopausal endometriosis to help inform clinical decision-making and point to novel approaches for treatment and management. RECENT FINDINGS Although one unifying theory to explain the pathogenesis of endometriotic lesions has not been elucidated, estrogen dependence is central to the pathophysiological process. The total quantity of estrogen production is mediated by multiple enzymes in complex pathways. Recent studies have confirmed the presence of these necessary enzymes in endometriotic lesions thereby suggesting a local source of estrogen and a likely pathogenic contributor. More research is needed to fully elucidate the mechanism of local estrogen biosynthesis; however, the current data provide possible explanations for the presence of postmenopausal endometriosis in an otherwise systemically hypoestrogenic environment. SUMMARY All suspected endometriosis lesions should be surgically excised for optimization of treatment and prevention of malignant transformation. If hormone replacement therapy is initiated, combined estrogen and progestin is recommended, even in the setting of previous hysterectomy, given the risk of disease reactivation and malignant transformation of endometriotic lesions. Further research is needed to understand the true prevalence, cause, and progression in this patient demographic. Histologic studies evaluating tissue lesions and peritoneal fluid for estrogen receptors, estrogen metabolizing enzymes, immune cells, and nerve fibers will aide in clinical management and treatment planning.
Collapse
|
27
|
Du Y, Zhang Z, Xiong W, Li N, Liu H, He H, Li Q, Liu Y, Zhang L. Estradiol promotes EMT in endometriosis via MALAT1/miR200s sponge function. Reproduction 2019; 157:179-188. [PMID: 30500775 PMCID: PMC7305834 DOI: 10.1530/rep-18-0424] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022]
Abstract
Endometriosis is an estrogen-dependent benign gynecological disease that shares some common features of malignancy. Epithelial-mesenchymal transition (EMT) has been recognized as a core mechanism of endometriosis. MALAT1 is widely known as EMT promoter, while miR200 family members (miR200s) are considered as EMT inhibitors. Previous studies have reported that MALAT1 upregulation and miR200s downregulation are observed in endometriosis. MiR200c has been regarded as the strongest member of miR200s to interact with MALAT1. However, whether MALAT1/miR200c regulates EMT remains largely unclear. In this study, the roles of miR200s and MALAT1 in ectopic endometrium were investigated. Additionally, the effects of E2 on EMT and MALAT1/miR200s were examined in both EECs and Ishikawa cells. Notably, E2 could upregulate MALAT1 and downregulate miR200s expression levels and induce EMT in EECs and Ishikawa cells. PHTPP, an ERβ antagonist, could reverse the effect of E2. Overexpression of miR200c and knockdown of MALAT1 significantly inhibited E2-mediated EMT, suggesting that both miR200c and MALAT1 are involved in the E2-induced EMT process in endometriosis. In addition, a reciprocal inhibition was found between miR200s and MALAT1. Therefore, the role of MALAT1/miR200c in EMT is influenced by the presence of estrogen during endometriosis development.
Collapse
Affiliation(s)
- Yu Du
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhibing Zhang
- Department of Physiology and Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Wenqian Xiong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Na Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hengwei Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haitang He
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ling Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
28
|
Heinosalo T, Gabriel M, Kallio L, Adhikari P, Huhtinen K, Laajala TD, Kaikkonen E, Mehmood A, Suvitie P, Kujari H, Aittokallio T, Perheentupa A, Poutanen M. Secreted frizzled-related protein 2 (SFRP2) expression promotes lesion proliferation via canonical WNT signaling and indicates lesion borders in extraovarian endometriosis. Hum Reprod 2019; 33:817-831. [PMID: 29462326 DOI: 10.1093/humrep/dey026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/24/2018] [Indexed: 12/21/2022] Open
Abstract
STUDY QUESTION What is the role of SFRP2 in endometriosis? SUMMARY ANSWER SFRP2 acts as a canonical WNT/CTNNB1 signaling agonist in endometriosis, regulating endometriosis lesion growth and indicating endometriosis lesion borders together with CTNNB1 (also known as beta catenin). WHAT IS KNOWN ALREADY Endometriosis is a common, chronic disease that affects women of reproductive age, causing pain and infertility, and has significant economic impact on national health systems. Despite extensive research, the pathogenesis of endometriosis is poorly understood, and targeted medical treatments are lacking. WNT signaling is dysregulated in various human diseases, but its role in extraovarian endometriosis has not been fully elucidated. STUDY DESIGN, SIZE, DURATION We evaluated the significance of WNT signaling, and especially secreted frizzled-related protein 2 (SFRP2), in extraovarian endometriosis, including peritoneal and deep lesions. The study design was based on a cohort of clinical samples collected by laparoscopy or curettage and questionnaire data from healthy controls and endometriosis patients. PARTICIPANTS/MATERIALS, SETTING, METHODS Global gene expression analysis in human endometrium (n = 104) and endometriosis (n = 177) specimens from 47 healthy controls and 103 endometriosis patients was followed by bioinformatics and supportive qPCR analyses. Immunohistochemistry, Western blotting, primary cell culture and siRNA knockdown approaches were used to validate the findings. MAIN RESULTS AND THE ROLE OF CHANCE Among the 220 WNT signaling and CTNNB1 target genes analysed, 184 genes showed differential expression in extraovarian endometriosis (P < 0.05) compared with endometrium tissue, including SFRP2 and CTNNB1. Menstrual cycle-dependent regulation of WNT genes observed in the endometrium was lost in endometriosis lesions, as shown by hierarchical clustering. Immunohistochemical analysis indicated that SFRP2 and CTNNB1 are novel endometriosis lesion border markers, complementing immunostaining for the known marker CD10 (also known as MME). SFRP2 and CTNNB1 localized similarly in both the epithelium and stroma of extraovarian endometriosis tissue, and interestingly, both also indicated an additional distant lesion border, suggesting that WNT signaling is altered in the endometriosis stroma beyond the primary border indicated by the known marker CD10. SFRP2 expression was positively associated with pain symptoms experienced by patients (P < 0.05), and functional loss of SFRP2 in extraovarian endometriosis primary cell cultures resulted in decreased cell proliferation (P < 0.05) associated with reduced CTNNB1 protein expression (P = 0.05). LIMITATIONS REASONS FOR CAUTION SFRP2 and CTNNB1 improved extraovarian endometriosis lesion border detection in a relatively small cohort (n = 20), although larger studies with different endometriosis subtypes in variable cycle phases and under hormonal medication are required. WIDER IMPLICATIONS OF THE FINDINGS The highly expressed SFRP2 and CTNNB1 improve endometriosis lesion border detection, which can have clinical implications for better visualization of endometriosis lesions over CD10. Furthermore, SFRP2 acts as a canonical WNT/CTNNB1 signaling agonist in endometriosis and positively regulates endometriosis lesion growth, suggesting that the WNT pathway may be an important therapeutic target for endometriosis. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the Academy of Finland and by Tekes: Finnish Funding Agency for Innovation. The authors have no conflict of interest to declare.
Collapse
Affiliation(s)
- T Heinosalo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland
| | - M Gabriel
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland.,Department of Obstetrics and Gynecology, University of Turku and Turku University Hospital, 20014 Turku, Finland
| | - L Kallio
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland
| | - P Adhikari
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland
| | - K Huhtinen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland.,Institute of Biomedicine, Research Center for Cancer, Infections and Immunity, University of Turku, 20014 Turku, Finland.,Department of Pathology, Turku University Hospital, 20521 Turku, Finland
| | - T D Laajala
- Department of Mathematics and Statistics, University of Turku, 20014 Turku, Finland.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland.,Turku Center for Disease Modeling (TCDM), University of Turku, 20014 Turku, Finland
| | - E Kaikkonen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland
| | - A Mehmood
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland
| | - P Suvitie
- Department of Obstetrics and Gynecology, University of Turku and Turku University Hospital, 20014 Turku, Finland
| | - H Kujari
- Institute of Biomedicine, Research Center for Cancer, Infections and Immunity, University of Turku, 20014 Turku, Finland.,Department of Pathology, Turku University Hospital, 20521 Turku, Finland
| | - T Aittokallio
- Department of Mathematics and Statistics, University of Turku, 20014 Turku, Finland.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland.,Turku Center for Disease Modeling (TCDM), University of Turku, 20014 Turku, Finland
| | - A Perheentupa
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland.,Department of Obstetrics and Gynecology, University of Turku and Turku University Hospital, 20014 Turku, Finland
| | - M Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, 20014 Turku, Finland.,Turku Center for Disease Modeling (TCDM), University of Turku, 20014 Turku, Finland.,Institute of Medicine, Sahlgrenska Academy, 405 30 Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
29
|
Zhang H, Li G, Sheng X, Zhang S. Upregulation of miR‑33b promotes endometriosis via inhibition of Wnt/β‑catenin signaling and ZEB1 expression. Mol Med Rep 2019; 19:2144-2152. [PMID: 30664209 PMCID: PMC6390049 DOI: 10.3892/mmr.2019.9870] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 07/30/2018] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the role and mechanisms of microRNA (miR)‑33b in endometriosis (Ems). Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR), MTT assays, flow cytometry, caspase‑3/9 activity assays and western blotting were performed in the present study. Initially, miR‑33b expression in an Ems rat model was investigated by RT‑qPCR and was demonstrated to be upregulated in Ems tissue samples of rats compared with the control group. In addition, miR‑33b upregulation inhibited cell growth and enhanced apoptosis in an Ems model (primary cell cultures) compared with the control group. In addition, miR‑33b up‑regulation reduced Wnt/β‑catenin signaling pathway and suppressed zinc finger E‑box‑binding homeobox 1 (ZEB1) protein expression in the in vitro Ems model (primary cell cultures) compared with the control group. Furthermore, small interfering‑ZEB1 ameliorated the effects of miR‑33b downregulation on Ems cell growth in the in vitro Ems model. Additionally, a Wnt agonist reduced the effects of miR‑33b upregulation on Ems cell growth in the in vitro Ems model. In conclusion, the present study demonstrated that upregulation of miR‑33b may promote Ems through Wnt/β‑catenin by ZEB1 expression.
Collapse
Affiliation(s)
- Haiyan Zhang
- Department of Gynecology, Affiliated Qilu Hospital of Shandong University, Jinan, Shandong 250002, P.R. China
| | - Guang Li
- Department of Gynecology Ward 1, Linyi City People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Xiugui Sheng
- Department of Gynecology, Chinese Academy of Medical Sciences Tumor Hospital, Beijing 100021, P.R. China
| | - Shiqian Zhang
- Department of Gynecology, Affiliated Qilu Hospital of Shandong University, Jinan, Shandong 250002, P.R. China
| |
Collapse
|
30
|
Ek M, Roth B, Engström G, Ohlsson B. AXIN1 in Plasma or Serum Is a Potential New Biomarker for Endometriosis. Int J Mol Sci 2019; 20:ijms20010189. [PMID: 30621017 PMCID: PMC6337238 DOI: 10.3390/ijms20010189] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/26/2018] [Accepted: 12/30/2018] [Indexed: 12/12/2022] Open
Abstract
Although endometriosis is considered an inflammatory disease, no reliable diagnostic biomarkers exist for use in clinical practice. The aim was to investigate the inflammatory profile in endometriosis using an exploratory approach of inflammation-related proteins. Patients with laparoscopy-verified endometriosis (N = 172), women with microscopic colitis (N = 50), healthy controls (N = 31), and age-matched controls from the general population (N = 100) were enrolled and questionnaires regarding socioeconomic factors, lifestyle habits, and medical history were completed. Sera from patients and healthy controls were analyzed for 92 inflammatory biomarkers using Proximity Extension Assay technology (PEA). Plasma AXIN1 levels were analyzed in patients with endometriosis and controls from the general population by ELISA. General linear model adjusted for age, Mann–Whitney U-test, and principal component analysis (PCA) were used for statistical calculations. Serum levels of AXIN1 and ST1A1 were increased in endometriosis compared with MC (p < 0.001) and healthy controls (p = 0.001), whereas CXCL9 levels were decreased. Plasma levels of AXIN1 were elevated in endometriosis compared with age-matched controls from the general population (30.0 (17.0–38.0) pg/mL vs. 19.5 (15.0–28.0) pg/mL, p < 0.001). PCA analysis identified four clusters of proteins, where one cluster differed between endometriosis and controls, with strong correlations for AXIN1 and ST1A1. Plasma/serum AXIN1 is an interesting biomarker to be further evaluated in endometriosis.
Collapse
Affiliation(s)
- Malin Ek
- Department of Internal Medicine, Skåne University Hospital, Lund University, 221 00 Lund, Sweden.
| | - Bodil Roth
- Department of Internal Medicine, Skåne University Hospital, Lund University, 221 00 Lund, Sweden.
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, Clinical Research Centre, Lund University, Box 50332, 202 13 Malmö, Sweden.
| | - Bodil Ohlsson
- Department of Internal Medicine, Skåne University Hospital, Lund University, 221 00 Lund, Sweden.
| |
Collapse
|
31
|
Mai H, Wei Y, Yin Y, Huang S, Lin H, Liao Y, Liu X, Chen X, Shi H, Liu C, Xu H. LINC01541 overexpression attenuates the 17β-Estradiol-induced migration and invasion capabilities of endometrial stromal cells. Syst Biol Reprod Med 2019; 65:214-222. [PMID: 30608887 DOI: 10.1080/19396368.2018.1549290] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hong Mai
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yeping Wei
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yan Yin
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shijin Huang
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huisi Lin
- Department of Gynaecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yan Liao
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xupeng Liu
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xianfeng Chen
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Haijuan Shi
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chuanzhong Liu
- Department of Gynaecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hong Xu
- Department of Gynaecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
32
|
Hu C, Zhen Y, Pang B, Lin X, Yi H. Myeloid-Derived Suppressor Cells Are Regulated by Estradiol and Are a Predictive Marker for IVF Outcome. Front Endocrinol (Lausanne) 2019; 10:521. [PMID: 31417498 PMCID: PMC6682648 DOI: 10.3389/fendo.2019.00521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
In vitro fertilization (IVF) is an effective means to treat infertility, but the pregnancy rate is still unsatisfactory and reliable markers to predict pregnancy outcome are ill-defined. Myeloid-derived suppressor cell (MDSC) are critically involved in decisions related to the acceptance or rejection of foreign fetal antigens by the maternal immune system. However, factors that regulate peripheral blood MDSC during pre-pregnancy are poorly defined. Thus, the goal of this study was to assess the relationships among serum estradiol (E2) and endothelial growth factor (VEGF) levels, MDSC ratios, and pregnancy outcome associated with IVF. Patients undergoing IVF treatment (n = 54) were recruited from January to June 2018. Levels of E2 and VEGF were measured by ELISA, MDSC ratios among peripheral blood mononuclear cells (PBMC) were detected by flow cytometry, and the crosstalk among these parameters was analyzed. A receiver operating characteristic curve (ROC) of MDSC levels was plotted to assess this measure as an independent predictive factor for pregnancy. In addition, we analyzed the possible involvement of molecular pathways by bioinformatics. When E2 levels were <4,000 pg/ml, MDSC proportion was positively correlated with serum E2 and VEGF levels. However, when E2 levels were >4,000 pg/ml, MDSC ratio and VEGF levels were negatively correlated with E2. A ROC curve revealed that the percentage of MDSC had better sensitivity and specificity at a concentration of 8.22% (0.875 and 0.75, respectively; area under the curve (AUC) = 0.859) to predict pregnancy success, based on multiple logistic regression analysis. Furthermore, we found 12 target genes of E2 and VEGF, and also functional genes related to MDSC, indicating potential protein-protein interactions underlying these associations. In summary, we showed that E2, depending on its concentration, might play a dichotomous role in influencing the MDSC proportion by regulating VEGF. In IVF patients, an increased MDSC ratio among PBMC was highly correlated with elevated pregnancy rates, independent of the effects of E2, which might provide new insight into immune-related miscarriage and IVF failure.
Collapse
Affiliation(s)
- Cong Hu
- Central Laboratory of the Eastern Division, The First Hospital of Jilin University, Changchun, China
- Center for Reproductive Medicine, Center for Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, China
| | - Yu Zhen
- Department of Dermatology, The First Hospital of Jilin University, Changchun, China
| | - Bo Pang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Xiuying Lin
- Center for Reproductive Medicine, Jilin Province People's Hospital, Changchun, China
| | - Huanfa Yi
- Central Laboratory of the Eastern Division, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Huanfa Yi
| |
Collapse
|
33
|
Khamphukdee C, Monthakantirat O, Chulikhit Y, Buttachon S, Lee M, Silva AMS, Sekeroglu N, Kijjoa A. Chemical Constituents and Antidepressant-Like Effects in Ovariectomized Mice of the Ethanol Extract of Alternanthera philoxeroides. Molecules 2018; 23:E2202. [PMID: 30200295 PMCID: PMC6225253 DOI: 10.3390/molecules23092202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 01/30/2023] Open
Abstract
The previously unreported flavone glycoside, demethyltorosaflavone B (2) and the E-propenoic acid substituted flavone, torosaflavone E (3a), were isolated together with nine previously reported metabolites, including indole-3-carbaldehyde, oleanonic acid, vanillic acid, p-hydroxybenzoic acid, altheranthin (1a), alternanthin B (1b), demethyltorosaflavone D (3b), luteolin 8-C-E-propenoic acid (4) and chrysoeriol 7-O-rhamnoside (5), from the ethanol extract of the aerial part of Althernanthera philoxeroides. The crude ethanol extract was evaluated for its in vitro estrogenic activity in MCF-7 breast cancer cell line. The crude ethanol extract was also investigated in vivo for its antidepressant-like effects on ovariectomized mice using tail suspension and forced swimming tests, while its effect on the locomotor activity was evaluated by a Y-maze test. The effect of the crude extract on the serum corticosterone level, size and volume of uterus of the ovariectomized mice were also investigated. The expression of the mouse cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF) and β-actin mRNAs in hippocampus and frontal cortex was also evaluated, using semiquantitative reverse transcription-polymerase chain reaction. The crude extract and the isolated compounds 1a, 1b, 3a, 3b and 5, were evaluated for their inhibitory effects on monoamine oxidases (MAOs)-A and -B.
Collapse
Affiliation(s)
- Charinya Khamphukdee
- Graduate School of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Orawan Monthakantirat
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Yaowared Chulikhit
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Suradet Buttachon
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto de Lexões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal.
| | - Michael Lee
- Department of Chemistry, University of Leicester, University Road, Leicester LE 7 RH, UK.
| | - Artur M S Silva
- Departamento de Química & QOPNA, Universidade de Aveiro, 3810-193 Aveiro, Portugal.
| | - Nazim Sekeroglu
- Department of Food Engineering, Faculty of Architecture and Engineering, Kilis 7 Aralık University, Kilis 79000, Turkey.
| | - Anake Kijjoa
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto de Lexões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal.
| |
Collapse
|
34
|
Klemmt PA, Starzinski-Powitz A. Molecular and Cellular Pathogenesis of Endometriosis. CURRENT WOMEN'S HEALTH REVIEWS 2018; 14:106-116. [PMID: 29861704 PMCID: PMC5925869 DOI: 10.2174/1573404813666170306163448] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 02/06/2017] [Accepted: 02/15/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND A substantial body of studies supports the view that molecular and cellular features of endometriotic lesions differ from those of eutopic endometrium. Apart from that, evidence exists that the eutopic endometrium from pa-tients with endometriosis differs from that of females without endometriosis. OBJECTIVE Aberrant expression profiles include a number of non-steroid signaling pathways that exert their putative influ-ence on the pathogenesis of endometriosis at least in part via crosstalk(s) with estrogen-mediated mechanisms. A rational to focus research on non-steroid signal pathways is that they might be remunerative targets for the development and selection of novel therapeutics to treat endometriosis possibly without affecting estrogen levels. RESULTS AND CONCLUSION In this article, we describe molecular and cellular features of endometriotic lesions and focus on the canonical WNT/β-signaling pathway, a key regulatory system in biology (including stem cell homeostasis) and often in pathophysiological conditions such as endometriosis. Recently emerged novel biological concepts in signal transduction and gene regulation like exosomes and microRNAs are discussed in their putative role in the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Petra A.B. Klemmt
- Department of Molecular Cell Biology and Human Genetics, Institute of Cell Biology and Neuroscience, Johann Wolfgang Goethe University of Frankfurt, Max-von-Laue-Str. 13, D-60438Frankfurt am Main, Germany
| | - Anna Starzinski-Powitz
- Department of Molecular Cell Biology and Human Genetics, Institute of Cell Biology and Neuroscience, Johann Wolfgang Goethe University of Frankfurt, Max-von-Laue-Str. 13, D-60438Frankfurt am Main, Germany
| |
Collapse
|
35
|
Yang YM, Yang WX. Epithelial-to-mesenchymal transition in the development of endometriosis. Oncotarget 2018; 8:41679-41689. [PMID: 28415639 PMCID: PMC5522315 DOI: 10.18632/oncotarget.16472] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/11/2017] [Indexed: 12/15/2022] Open
Abstract
Endometriosis, an estrogen-dependent chronic gynecological disease, is common in reproductive-age women and profoundly affects their life quality. Although various pathogenic theories have been proposed, the origin of endometriosis remains unclear. Epithelial to mesenchymal transition (EMT) is a process that epithelial cells lose polarized organization of the cytoskeleton and cell-to-cell contacts, acquiring the high motility of mesenchymal cells. These changes are thought to be prerequisites for the original establishment of endometriotic lesions. However, no study exactly indicates which type of EMT occurs in endometriosis. In this review, we conclude that two different types of EMT may participate in this disease. Besides, two stimulating signals, hypoxia and estrogen, can through different pathways to activate the EMT process in endometriosis. Those pathways involve many cellular factors such as TGF-beta and Wnt, ultimately leading to cell proliferation and migration. As infertility is becoming a serious and intractable issue for women, EMT, during the implantation process, is gaining attention. In this review, we will describe the known functions of EMT in endometriosis, and suggest further studies that may aid in the development of medical therapy.
Collapse
Affiliation(s)
- Yan-Meng Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
36
|
Liu H, Tao Y, Chen M, Yu J, Li WJ, Tao L, Li Y, Li F. 17β-Estradiol Promotes Angiogenesis of Rat Cardiac Microvascular Endothelial Cells In Vitro. Med Sci Monit 2018; 24:2489-2496. [PMID: 29684003 PMCID: PMC5936052 DOI: 10.12659/msm.903344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The formation of new blood vessels, known as angiogenesis, is critical for recovery from ischemic heart disease, and estrogen is considered an important factor in this process. Here, we investigated the effects of 17β-estradiol (17β-E2) on proliferation and migration of cardiac microvascular endothelial cells (CMECs) in vitro. Material/Methods Rat CMECs were isolated and cultured with 17β-E2 (0.001–1 μmol/l) in the absence or presence of the estrogen antagonist tamoxifen. Then, the expression level of estrogen receptor alpha was evaluated by using immunofluorescence assay, RT-PCR, and Western blot. Cell proliferation was detected by methyl thiazolyl tetrazolium analysis and the cell migration was verified by a scraping assay and quantified by a Transwell chamber assay. CMEC differentiation was examined using a tube formation assay. Vascular endothelial growth factor (VEGF) secretion was detected by enzyme-linked immunosorbent assay. Results CMECs exhibited homogenous, polygonal, exhibited contact inhibition, and had characteristically ovoid nuclei with 1 or 2 nucleoli, and the cytoplasm exhibited red fluorescence after staining for von Willebrand factor. 17β-E2 treatment upregulated estrogen receptor alpha expression in CMECs. 17β-E2 treatment significantly promoted the proliferation, migration, tubular structure formation, and VEGF secretion in CMECs. The maximal proliferation occurred in the presence of 0.01 μmol/l 17β-E2. Furthermore, estrogen and VEGF were found to synergistically stimulate angiogenesis. Conclusions Our data show that 17β-E2 promotes angiogenesis in vitro and suggests that estrogen treatment as a novel therapeutic modality in the management of arterial insufficiency.
Collapse
Affiliation(s)
- HaiTao Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Yin Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Mai Chen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Jin Yu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Wei-Jie Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Yan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Fei Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
37
|
Wu F, Wu L, Wu Q, Zhou L, Li W, Wang D. Duplication and gene expression patterns of β-catenin in Nile tilapia. FISH PHYSIOLOGY AND BIOCHEMISTRY 2018; 44:651-659. [PMID: 29290067 DOI: 10.1007/s10695-017-0460-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 12/18/2017] [Indexed: 06/07/2023]
Abstract
β-Catenin, a key transcriptional coactivator of the Wnt pathway, plays an important role in animal embryonic development and organogenesis. In our earlier study, we have reported that two types of β-catenin (β-catenin-1 and β-catenin-2) were ubiquitously expressed in almost all the tissues examined in tilapia. However, the immunolocalization of β-catenin in those tissues, especially in extra-gonadal tissues, remains unclear. In the present study, we further confirm that these two types of β-catenin gene exist only in teleosts and are derived from 3R (third round of genome duplication) by phylogenetic and syntenic analyses. Moreover, the transcriptome analysis conducted in this investigation reveals that two β-catenins exhibited similar expression patterns in seven adult tissues and four key developmental stages of XX and XY gonads. Finally, immunohistochemistry analysis was performed to detect the cell localization of β-catenin. A positive signal of β-catenin was observed in various tissues of tilapia, such as the intestine, liver, kidney, spleen, eye, brain, and gonads. The results of our study indicate that tilapia β-catenin might be involved in the organ development and play some specific functions in biological processes; all these data will provide basic reference for understanding the molecular mechanism of β-catenin in regulating of teleost organogenesis.
Collapse
Affiliation(s)
- Fengrui Wu
- Key Laboratory of Embryo Development and Reproductive Regulation, School of Biological and Food Engineering, Fuyang Teachers College, Fuyang, Anhui Province, 236000, China.
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing, 400715, China.
| | - Limin Wu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing, 400715, China
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Qingqing Wu
- Key Laboratory of Embryo Development and Reproductive Regulation, School of Biological and Food Engineering, Fuyang Teachers College, Fuyang, Anhui Province, 236000, China
| | - Linyan Zhou
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing, 400715, China
| | - Wenyong Li
- Key Laboratory of Embryo Development and Reproductive Regulation, School of Biological and Food Engineering, Fuyang Teachers College, Fuyang, Anhui Province, 236000, China
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing, 400715, China
| |
Collapse
|
38
|
Li N, Zhang L, Li Q, Du Y, Liu H, Liu Y, Xiong W. Notch activity mediates oestrogen-induced stromal cell invasion in endometriosis. Reproduction 2018; 157:371-381. [PMID: 30753135 DOI: 10.1530/rep-18-0326] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 02/11/2019] [Indexed: 01/04/2023]
Abstract
Oestrogen has been reported to control the invasiveness of endometrial stromal cells in endometriosis. Notch signalling, a master regulator of cell invasion in tumours, is regulated by oestrogen in other diseases and hyperactivated in endometriotic stromal cells. Therefore, we hypothesized that an interaction between Notch signalling and oestrogen may exist in the regulation of endometrial stromal cell invasion, which is essential for the development of endometriosis. Western blot analysis of tissues showed that the expression levels of Notch components (JAG1 and NOTCH1) and Notch activity were markedly higher in ectopic endometria than in their eutopic and normal counterparts. Primary stromal cells obtained from normal endometria cultured with oestrogen presented significant increases in the expression of Notch components and Notch activity, the cytoplasmic and nuclear accumulation of NOTCH1 intracellular domain, the expression of matrix metallopeptidase 9 and vascular endothelial growth factor and cell invasiveness. Knockdown ofNOTCH1markedly alleviated oestrogen-induced matrix metallopeptidase 9 and vascular endothelial growth factor expression and cell invasion. ICI (an oestrogen receptor α antagonist) also blocked these oestrogenic effects. Oestrogen-responsive elements were found in the promoters ofNOTCH1andJAG1. A luciferase reporter analysis revealed that oestrogen regulated the expression of Notch components via oestrogen receptor alpha, which is bound to oestrogen-responsive elements in theJAG1andNOTCH1promoters. Collectively, our findings indicate that oestrogen engages in crosstalk with Notch signalling to regulate cell invasion in endometriosis via the activation of oestrogen receptor alpha and the enhancement of Notch activity. Notch signalling blockade may therefore be a novel therapeutic target for endometriosis.
Collapse
Affiliation(s)
- Na Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Du
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hengwei Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenqian Xiong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
39
|
Abstract
Oestrogen–progesterone signalling is highly versatile and critical for the maintenance of healthy endometrium in humans. The genomic and nongenomic signalling cascades initiated by these hormones in differentiated cells of endometrium have been the primary focus of research since 1920s. However, last decade of research has shown a significant role of stem cells in the maintenance of a healthy endometrium and the modulatory effects of hormones on these cells. Endometriosis, the growth of endometrium outside the uterus, is very common in infertile patients and the elusiveness in understanding of disease pathology causes hindrance in selection of treatment approaches to enhance fertility. In endometriosis, the stem cells are dysfunctional as it can confer progesterone resistance to their progenies resulting in disharmony of hormonal orchestration of endometrial homeostasis. The bidirectional communication between stem cell signalling pathways and oestrogen–progesterone signalling is found to be disrupted in endometriosis though it is not clear which precedes the other. In this paper, we review the intricate connection between hormones, stem cells and the cross-talks in their signalling cascades in normal endometrium and discuss how this is deregulated in endometriosis. Re-examination of the oestrogen–progesterone dependency of endometrium with a focus on stem cells is imperative to delineate infertility associated with endometriosis and thereby aid in designing better treatment modalities.
Collapse
|
40
|
Zhang G, Li C, Zhu N, Chen Y, Yu Q, Liu E, Wang R. Sex differences in the formation of atherosclerosis lesion in apoE-/-mice and the effect of 17β-estrodiol on protein S-nitrosylation. Biomed Pharmacother 2018; 99:1014-1021. [DOI: 10.1016/j.biopha.2018.01.145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/12/2018] [Accepted: 01/28/2018] [Indexed: 01/19/2023] Open
|
41
|
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: 7.9] [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
|
42
|
Liu Y, Ma Y. Promoter Methylation Status of WNT2 in Placenta from Patients with Preeclampsia. Med Sci Monit 2017; 23:5294-5301. [PMID: 29109390 PMCID: PMC5687122 DOI: 10.12659/msm.903602] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Preeclampsia is a serious multisystem disorder of human gestation, affecting up to 10% of pregnant women worldwide, and results in maternal morbidity and mortality. The aim of this study was to determine the gene expression pattern and methylation status of the promoter of the WNT2 gene in placentas from patients with preeclampsia and to evaluate the potential role of the WNT2 pathway in the pathogenesis of preeclampsia. Material/Methods Real-time quantitative polymerase chain reaction (PT-PCR) was used to determine the WNT2 gene expression level. Western blot analysis was used to identify alterations in wnt2 protein expression. Results The mRNA and protein expression levels of the WNT2 gene were reduced in placentas from patients with preeclampsia when compared with placentas from healthy women. The average methylation level of the promoter of the WNT2 gene was elevated in the placentas from patients with preeclampsia compared with the controls placentas from healthy women. Conclusions The findings of this study have shown that molecular mechanisms, including aberrant activation of the WNT2 gene signaling pathway, may be involved in the pathogenesis of preeclampsia. Promoter hypermethylation and reduced expression of the WNT2 gene requires further study to determine a potential role in the diagnosis and treatment of preeclampsia.
Collapse
Affiliation(s)
- Yufang Liu
- Department of Gynaecology and Obstetrics, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland).,Department of Gynaecology and Obstetrics, Binzhou Medical University Hospital, Binzhou, Shandong, China (mainland)
| | - Yuyan Ma
- Department of Gynaecology and Obstetrics, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
43
|
Cakmak H, Seval-Celik Y, Arlier S, Guzeloglu-Kayisli O, Schatz F, Arici A, Kayisli UA. p38 Mitogen-Activated Protein Kinase is Involved in the Pathogenesis of Endometriosis by Modulating Inflammation, but not Cell Survival. Reprod Sci 2017; 25:587-597. [PMID: 28845752 DOI: 10.1177/1933719117725828] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Local pro-inflammatory environment and enhanced cell survival contribute to the endometriosis development. A serine/threonine kinase p38 mitogen-activated protein kinase (MAPK) mediates intracellular signaling of cytokine production, cell proliferation, and apoptosis in different cell types. The current study compares p38 MAPK activity in normal endometrium and endometriosis, and assesses role(s) of p38 MAPK on cytokine production and cell survival in endometriosis. METHODS Immunohistochemical levels of total and phosphorylated (active) p38 MAPK as well as its correlation with interleukin 8 (IL-8) expression, and cell proliferation and apoptosis were compared in normal human endometrium and endometriosis. The action of p38 MAPK on pro-inflammatory cytokine-induced IL-8 and monocyte chemotactic protein (MCP)-1 expression in endometriotic cells were assessed by enzyme-linked immunosorbent assay. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide cell survival, 5-bromo-2'-deoxyuridine incorporation, and Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling assays were used to determine the function of p38 MAPK in cultured human endometriotic stromal cell proliferation and apoptosis. RESULTS p38 MAPK activity was significantly higher in both eutopic and ectopic endometria compared to normal endometria during late proliferative and early secretory phases ( P < .05). Increased p38 MAPK activity in endometriotic cells correlated with IL-8 expression (Pearson correlation coefficient r = 0.83, P < .01), but not with apoptosis in vivo. The pro-inflammatory cytokines IL-1β and tumor necrosis factor (TNF)-α induced activation of p38 MAPK. Inhibition of p38 MAPK activity blocked IL-1β and TNF-α-induced IL-8 and MCP-1 secretion in cultured endometriotic stromal cells ( P < .05), but did not impact on endometriotic cell survival. CONCLUSIONS These results suggest that rather than modulating cell survival, increased p38 MAPK activity in endometriotic cells contributes to the pathogenesis of endometriosis by promoting the local inflammatory milieu.
Collapse
Affiliation(s)
- Hakan Cakmak
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.,2 Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yasemin Seval-Celik
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.,3 Department of Histology and Embryology, Faculty of Medicine, Izmir University of Economics, Izmir, Turkey
| | - Sefa Arlier
- 4 Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Ozlem Guzeloglu-Kayisli
- 4 Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Frederick Schatz
- 4 Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Aydin Arici
- 1 Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Umit A Kayisli
- 4 Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
44
|
Li KL, Wang YF, Qin JR, Wang F, Yang YT, Zheng LW, Li MH, Kong J, Zhang W, Yang HY. Rapamycin enhances the anti-angiogenesis and anti-proliferation ability of YM155 in oral squamous cell carcinoma. Tumour Biol 2017; 39:1010428317706213. [PMID: 28618939 DOI: 10.1177/1010428317706213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
YM155, a small molecule inhibitor of survivin, has been studied in many tumors. It has been shown that YM155 inhibited oral squamous cell carcinoma through promoting apoptosis and autophagy and inhibiting proliferation. It was found that YM155 also inhibited the oral squamous cell carcinoma-mediated angiogenesis through the inactivation of the mammalian target of rapamycin pathway. Rapamycin, a mammalian target of rapamycin inhibitor, played an important role in the proliferation and angiogenesis of oral squamous cell carcinoma cell lines. In our study, cell proliferation assay, transwell assay, tube formation assay, and western blot assay were used to investigate the synergistic effect of rapamycin on YM155 in oral squamous cell carcinoma. Either in vitro or in vivo, rapamycin and YM155 exerted a synergistic effect on the inhibition of survivin and vascular endothelial growth factor through mammalian target of rapamycin pathway. Overall, our results revealed that low-dose rapamycin strongly promoted the sensitivity of oral squamous cell carcinoma cell lines to YM155.
Collapse
Affiliation(s)
- Kong-Liang Li
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yu-Fan Wang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jia-Ruo Qin
- 2 Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Feng Wang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yong-Tao Yang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Li-Wu Zheng
- 3 Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Ming-Hua Li
- 4 Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jie Kong
- 5 Department of Stomatology, Shenzhen Children's Hospital, Shenzhen, China
| | - Wei Zhang
- 6 Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Hong-Yu Yang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
45
|
Sritharen Y, Enriquez-Sarano M, Schaff HV, Casaclang-Verzosa G, Miller JD. Pathophysiology of Aortic Valve Stenosis: Is It Both Fibrocalcific and Sex Specific? Physiology (Bethesda) 2017; 32:182-196. [PMID: 28404735 PMCID: PMC6148342 DOI: 10.1152/physiol.00025.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/10/2017] [Accepted: 02/10/2017] [Indexed: 12/24/2022] Open
Abstract
Our understanding of the fundamental biology and identification of efficacious therapeutic targets in aortic valve stenosis has lagged far behind the fields of atherosclerosis and heart failure. In this review, we highlight the most clinically relevant problems facing men and women with fibrocalcific aortic valve stenosis, discuss the fundamental biology underlying valve calcification and fibrosis, and identify key molecular points of intersection with sex hormone signaling.
Collapse
Affiliation(s)
- Yoginee Sritharen
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
| | | | - Hartzell V Schaff
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
| | - Grace Casaclang-Verzosa
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Jordan D Miller
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota;
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Department of Surgery, Mayo Clinic, Rochester, Minnesota; and the
- Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
46
|
Syndecan-1 Acts as an Important Regulator of CXCL1 Expression and Cellular Interaction of Human Endometrial Stromal and Trophoblast Cells. Mediators Inflamm 2017; 2017:8379256. [PMID: 28293067 PMCID: PMC5331292 DOI: 10.1155/2017/8379256] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/22/2016] [Accepted: 01/19/2017] [Indexed: 01/25/2023] Open
Abstract
Successful implantation of the embryo into the human receptive endometrium is substantial for the establishment of a healthy pregnancy. This study focusses on the role of Syndecan-1 at the embryo-maternal interface, the multitasking coreceptor influencing ligand concentration, release and receptor presentation, and cellular morphology. CXC motif ligand 1, being involved in chemotaxis and angiogenesis during implantation, is of special interest as a ligand of Syndecan-1. Human endometrial stromal cells with and without Syndecan-1 knock-down were decidualized and treated with specific inhibitors to evaluate signaling pathways regulating CXC ligand 1 expression. Western blot analyses of MAPK and Wnt members were performed, followed by analysis of spheroid interactions between human endometrial cells and extravillous trophoblast cells. By mimicking embryo contact using IL-1β, we showed less ERK and c-Jun activation by depletion of Syndecan-1 and less Frizzled 4 production as part of the canonical Wnt pathway. Additionally, more beta-catenin was phosphorylated and therefore degraded after depletion of Syndecan-1. Secretion of CXC motif ligand 1 depends on MEK-1 with respect to Syndecan-1. Regarding the interaction of endometrial and trophoblast cells, the spheroid center-to-center distances were smaller after depletion of Syndecan-1. Therefore, Syndecan-1 seems to affect signaling processes relevant to signaling and intercellular interaction at the trophoblast-decidual interface.
Collapse
|
47
|
Zhang L, Xiong W, Li N, Liu H, He H, Du Y, Zhang Z, Liu Y. Estrogen stabilizes hypoxia-inducible factor 1α through G protein-coupled estrogen receptor 1 in eutopic endometrium of endometriosis. Fertil Steril 2016; 107:439-447. [PMID: 27939762 DOI: 10.1016/j.fertnstert.2016.11.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/08/2016] [Accepted: 11/08/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate whether G protein-coupled estrogen receptor (GPER, also known as GPR30 and GPER1) stabilizes hypoxia-inducible factor 1α (HIF-1α) in eutopic endometrium (EuEM) of endometriosis. DESIGN Immunohistochemical analysis and experimental in vitro study. SETTING University hospital. PATIENT(S) Patients with or without endometriosis. INTERVENTION(S) The EuEM and normal control endometrium (CoEM) were obtained by curettage. Primary cultured endometrial stromal cells (ESCs) were treated with 17β-E2, G1, or G15. MAIN OUTCOME MEASURE(S) The EuEM and CoEM were collected for immunohistochemistry. Western blot, polymerase chain reaction, ELISA, and dual luciferase experiments were used to detect expression of GPER, HIF-1α, vascular endothelial growth factor (VEGF), and matrix metalloproteinase 9 (MMP9) in ESCs. Estradiol and G1 were used as agonists of GPER, G15 as an antagonist. Migration of ESCs and endothelial tube formation of human umbilical vein endothelial cells cultured in medium collected from ESCs were measured. RESULT(S) Protein levels of GPER and HIF-1α were higher in EuEM than in CoEM. Protein levels of HIF-1α but not HIF-1α mRNA levels increased concurrently with GPER after E2 and G1 treatment. Furthermore, expression and activity of VEGF and MMP9 increased under E2 and G1 stimulation. However, these effects disappeared when GPER was blocked. CONCLUSION(S) G protein-coupled estrogen receptor stabilizes HIF-1α and thus promotes HIF-1α-induced VEGF and MMP9 in ESCs, which play critical roles in endometriosis.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, People's Republic of China
| | - Wenqian Xiong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, People's Republic of China
| | - Na Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, People's Republic of China
| | - Hengwei Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, People's Republic of China
| | - Haitang He
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, People's Republic of China
| | - Yu Du
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, People's Republic of China
| | - Zhibing Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia
| | - Yi Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, People's Republic of China.
| |
Collapse
|
48
|
Activin A Stimulates Aromatase via the ALK4-Smad Pathway in Endometriosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5791510. [PMID: 27833918 PMCID: PMC5090068 DOI: 10.1155/2016/5791510] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 08/17/2016] [Indexed: 01/13/2023]
Abstract
Endometriosis is an estrogen-dependent disease. We previously found that the expression of Activin A was upregulated in the peritoneal fluid of patients with endometriosis. The results of the present study indicated that Activin A induced estradiol secretion and P450arom expression in endometrial stromal cells (ESCs) derived from endometriosis patients. The mechanism of estrogenic synthesis was regulated by the Activin-Smad pathway in endometrial lesions. The data showed that the effect of Activin A on ESCs was partially abrogated by pretreatment with an inhibitor of ALK4 (the type I receptor, ActRIB) and Smad4-siRNA. Cumulatively, these data suggest that Activin A promotes the secretion of estradiol from ESCs by increasing the expression of P450arom via the ALK4-Smad pathway. These findings indicate the ALK4-Smad pathway may promote ectopic lesion survival and development.
Collapse
|
49
|
Huang C, Dong H, Zou M, Luo L, Hu Y, Xie Z, Le Y, Liu L, Zou F, Cai S. Bevacizumab reduced auto-phosphorylation of VEGFR2 to protect HDM-induced asthma mice. Biochem Biophys Res Commun 2016; 478:181-186. [PMID: 27453339 DOI: 10.1016/j.bbrc.2016.07.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/18/2016] [Indexed: 01/29/2023]
Abstract
Vascular endothelial growth factor (VEFG) is a major angiogenic factor involved in both normal physiological processes, such as embryonic development and wound healing, and in diseases, like cancer. Recent studies have revealed the functions of VEGF in inflammation and immunoregulation. Asthma is a chronic inflammation of the airways characterized by airway epithelial barrier dysfunction and imbalance in T-helper (Th) 1/Th2 during immunoregulation. We hypothesized that VEGF plays an important role in asthma. Utilizing a house dust mite extract (HDM)-induced murine model of asthma, we investigated whether bevacizumab, a humanized anti-VEGF monoclonal antibody, could protect the epithelial barrier in murine airways. We found that bevacizumab reduced airway hyper-responsiveness (AHR) and airway inflammation induced by HDM. In addition, HDM exposure promoted expression of VEGF, and caused AHR, disruptions of the epithelial barrier, and airway inflammation. Bevacizumab ameliorated AHR and the release of Th2 cytokines, thereby protecting the epithelial barrier. Our data suggest that bevacizumab may be a new therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Chaowen Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Mengchen Zou
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lishan Luo
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yahui Hu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhefan Xie
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanqing Le
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Laiyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fei Zou
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|