1
|
Yang Y, Li LL, Qi YX, Liu DJ. Research Progress of Caspase in Endometriosis. Reprod Sci 2024; 31:1496-1507. [PMID: 38347381 DOI: 10.1007/s43032-023-01425-3] [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: 09/18/2023] [Accepted: 12/08/2023] [Indexed: 05/24/2024]
Abstract
Endometriosis, a common chronic gynecological disease, refers to the presence and proliferation of endometrial tissue in locations other than the uterine cavity. Approximately 6 to 10% of the population of women of childbearing age are known to have endometriosis; the most common clinical signs are pelvic pain and infertility. Although endometriosis is a benign disease, it exhibits some typical features of malignant tumors, such as proliferation, invasion, metastasis, and recurrence. Endometriosis is considered a chronic, inflammatory, and estrogen-dependent disease, and multiple factors contribute to its occurrence and development. In recent years, increasing attention has been given to the role of apoptosis in the pathogenesis of this disease. Some researchers believe that spontaneous apoptosis of the endometrium is critical in maintaining its normal structure and function, and abnormal apoptosis can promote the occurrence and development of endometriosis. Inflammation is another likely process in the pathogenesis of endometriosis. Inflammation mediates the adhesion, proliferation, differentiation, and invasion of ectopic lesions of endometriosis, primarily by regulating the function of immune cells and increasing the level of proinflammatory cytokines in body fluids. The ultimate initiators of apoptosis and inflammatory cell death (pyroptosis) are the caspase family proteases. In this article, we review the progress in recent years in caspase function as well as the possible role of these enzymes in the pathogenesis of endometriosis, indicating potential treatment strategies.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Centre for Reproductive Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Lei-Lei Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Yu-Xin Qi
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Da-Jiang Liu
- Department of Gynecology and Obstetrics, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
2
|
Effect of Neiyi Prescription of QIU on autophagy and angiogenic ability of endometriosis via the PPARγ/NF-κB signaling pathway. Arch Gynecol Obstet 2022; 306:533-545. [DOI: 10.1007/s00404-022-06537-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 03/16/2022] [Indexed: 11/02/2022]
|
3
|
di Clemente N, Racine C, Pierre A, Taieb J. Anti-Müllerian Hormone in Female Reproduction. Endocr Rev 2021; 42:753-782. [PMID: 33851994 DOI: 10.1210/endrev/bnab012] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Joëlle Taieb
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| |
Collapse
|
4
|
Kaushal JB, Shukla V, Sankhwar P, Jha RK, Dwivedi A. Targeted inhibition of TAK1 abrogates TGFβ1 non-canonical signaling axis, NFκB/Smad7 inhibiting human endometriotic cells proliferation and inducing cell death involving autophagy. Cytokine 2021; 148:155700. [PMID: 34560609 DOI: 10.1016/j.cyto.2021.155700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022]
Abstract
Transforming growth factor (TGFβ) is known to play a major role in establishment and maintenance of endometriosis as reported by our group earlier, the underlying mechanism remains to be explored. We deciphered the involvement of TAK1 in TGFβ1- induced cellular responses and delineated the signaling mechanism in human endometriotic cells. The endometriotic cells showed elevated expression of TGFβ1 signaling-effector molecules. TGFβ1 exposure to endometriotic cells induced the expression of the downstream target molecules indicating that TGFβ1 is implicated in the commencement ofTAK1/NFκB-p65/Smad7 cascade. The silencing of TAK1 in endometriotic cells attenuated the TGFβ1 -induced NFκB transcriptional activation and nuclear translocation of NFκB-p65 subunit. The pharmacological inhibition of NFκB by QNZ or knockdown of TAK1 reduced the expression of Smad7 and Cox2. The knockdown of TAK1 in endometriotic cells showed G1 phase cell-cycle arrest and showed low BrdU-incorporation in the presence of TGFβ1. The inhibition of TAK1 attenuated the TGFβ1 signaling activation indicating that TAK1 is a crucial mediator for TGFβ1 action in endometriotic cells. The exposure of endometriotic cells to TAK1 inhibitor, celastrol caused activation of caspase-3 and -9 that led to PARP cleavage and induced apoptosis. Simultaneously, autophagy occurred in celastrol-treated and TAK1-silenced cells as was evidenced by the formation of autophagosome and the increased expression of autophagic markers. Thus, TAK1 activation appears to protect the growth of endometriotic cells by suppressing the cell death process. Overall, our study provided the evidence that of TAK1 significant in the endometriotic cell regulation and mediates a functional cross-talk between TGFβ1 and NFκB-p65 that promotes the growth and inflammatory response in endometriotic cells.
Collapse
Affiliation(s)
- Jyoti B Kaushal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), CSIR-CDRI Campus, Lucknow 226031, U.P., India
| | - Vinay Shukla
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), CSIR-CDRI Campus, Lucknow 226031, U.P., India
| | - Pushplata Sankhwar
- Department of Obstetrics & Gynaecology, King George's Medical University, Lucknow 226001, U.P., India
| | - Rajesh K Jha
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), CSIR-CDRI Campus, Lucknow 226031, U.P., India.
| | - Anila Dwivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), CSIR-CDRI Campus, Lucknow 226031, U.P., India.
| |
Collapse
|
5
|
Hung SW, Zhang R, Tan Z, Chung JPW, Zhang T, Wang CC. Pharmaceuticals targeting signaling pathways of endometriosis as potential new medical treatment: A review. Med Res Rev 2021; 41:2489-2564. [PMID: 33948974 PMCID: PMC8252000 DOI: 10.1002/med.21802] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 12/23/2020] [Accepted: 03/19/2021] [Indexed: 12/13/2022]
Abstract
Endometriosis (EM) is defined as endometrial tissues found outside the uterus. Growth and development of endometriotic cells in ectopic sites can be promoted via multiple pathways, including MAPK/MEK/ERK, PI3K/Akt/mTOR, NF-κB, Rho/ROCK, reactive oxidative stress, tumor necrosis factor, transforming growth factor-β, Wnt/β-catenin, vascular endothelial growth factor, estrogen, and cytokines. The underlying pathophysiological mechanisms include proliferation, apoptosis, autophagy, migration, invasion, fibrosis, angiogenesis, oxidative stress, inflammation, and immune escape. Current medical treatments for EM are mainly hormonal and symptomatic, and thus the development of new, effective, and safe pharmaceuticals targeting specific molecular and signaling pathways is needed. Here, we systematically reviewed the literature focused on pharmaceuticals that specifically target the molecular and signaling pathways involved in the pathophysiology of EM. Potential drug targets, their upstream and downstream molecules with key aberrant signaling, and the regulatory mechanisms promoting the growth and development of endometriotic cells and tissues were discussed. Hormonal pharmaceuticals, including melatonin, exerts proapoptotic via regulating matrix metallopeptidase activity while nonhormonal pharmaceutical sorafenib exerts antiproliferative effect via MAPK/ERK pathway and antiangiogenesis activity via VEGF/VEGFR pathway. N-acetyl cysteine, curcumin, and ginsenoside exert antioxidant and anti-inflammatory effects via radical scavenging activity. Natural products have high efficacy with minimal side effects; for example, resveratrol and epigallocatechin gallate have multiple targets and provide synergistic efficacy to resolve the complexity of the pathophysiology of EM, showing promising efficacy in treating EM. Although new medical treatments are currently being developed, more detailed pharmacological studies and large sample size clinical trials are needed to confirm the efficacy and safety of these treatments in the near future.
Collapse
Affiliation(s)
- Sze Wan Hung
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
| | - Ruizhe Zhang
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou
| | - Zhouyurong Tan
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
| | | | - Tao Zhang
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
| | - Chi Chiu Wang
- Department of Obstetrics and GynaecologyThe Chinese University of Hong KongHong Kong
- Reproduction and Development, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong
- School of Biomedical SciencesThe Chinese University of Hong KongHong Kong
- Chinese University of Hong Kong‐Sichuan University Joint Laboratory in Reproductive MedicineThe Chinese University of Hong KongHong Kong
| |
Collapse
|
6
|
The Multifaceted Role of Autophagy in Endometrium Homeostasis and Disease. Reprod Sci 2021; 29:1054-1067. [PMID: 33877643 DOI: 10.1007/s43032-021-00587-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
Autophagy is a conserved fundamental cellular process with a primary function of catabolizing harmful or surplus cellular contents such as protein aggregates, dysfunctional/long-lived organelles, intracellular pathogens, and storage nutrients. An increasing body of evidence reveals that basal autophagy is essential for maintaining endometrial homeostasis and mediating endometrial-specific functions, including menstrual cycle, embryo implantation, and decidualization. However, perturbed levels of autophagy can lead to severe endometrial pathologies, including endometriosis, endometrial hyperplasia, endometrial cancer, adenomyosis, and leiomyoma. This review highlights the most recent findings on the activity, regulation, and function of autophagy in endometrium physiology and pathology. Understanding the mechanistic roles of autophagy in endometrium homeostasis and disease is key to developing novel therapeutic strategies for endometrium-related infertility and malignancies.
Collapse
|
7
|
Luo X, Cheng W, Wang S, Chen Z, Tan J. Autophagy Suppresses Invasiveness of Endometrial Cells through Reduction of Fascin-1. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8615435. [PMID: 29992166 PMCID: PMC6016225 DOI: 10.1155/2018/8615435] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/09/2018] [Accepted: 04/26/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Autophagy has been reported to be involved in the development of various disorders such as neurodegenerative and metabolic diseases and tumors. Autophagy activators and inhibitors are also potential therapeutics for these diseases. However, the mechanism of autophagic involvement in different diseases is not the same, and the role of autophagy in endometriosis (EM) has not yet been elucidated. This research investigated the mechanism by which autophagy acts in EM, with the aim of establishing a theoretical basis for its prevention and treatment through the targeted interference with autophagy. METHODS We used an RNA interference fragment targeting ATG5, the autophagy activator rapamycin, and the autophagy inhibitor 3-MA or overexpression of filopodia-related protein fascin-1, in conjunction with clonogenic assays, growth curves, and scratch assay to investigate the influence of autophagy on cellular growth, proliferation, and invasiveness. We collected specimens from 20 clinical cases of EM and investigated the protein expression of the autophagic marker LC3-II, the autophagic substrate p62, and fascin-1. RESULTS Rapamycin was able to inhibit the proliferation and colony formation of the endometriotic cell line CRL-7566, whereas the autophagy inhibitor 3-MA as well as the interference with the autophagy-related gene ATG5 had the opposite effect. More importantly, the autophagy activator rapamycin was able to inhibit the growth of filopodia in the endometriotic cells, and the overexpression of the fascin-1 restored the rapamycin-induced decrease of invasiveness. We found that the expression of the autophagy marker LC3-II was significantly reduced among the clinical EM specimens compared to the control group, while the expressions of fascin-1 and autophagic substrate p62 were increased. CONCLUSION Our results indicate that the inhibition of autophagy and exogenous expression of fascin-1 may promote the invasiveness of endometrial cells. As a corollary, autophagy represents a potential target for the treatment of EM.
Collapse
Affiliation(s)
- Xiaomei Luo
- Department of Obstetrics and Gynecology, The Maternal and Child Healthcare Hospital of Hunan Province, Changsha 410005, China
| | - Wei Cheng
- Department of Obstetrics and Gynecology, The Maternal and Child Healthcare Hospital of Hunan Province, Changsha 410005, China
| | - Shizhang Wang
- Department of Obstetrics and Gynecology, The Maternal and Child Healthcare Hospital of Hunan Province, Changsha 410005, China
| | - Zhihong Chen
- Department of Pathology, The People's Hospital of Hunan Province, Changsha 410005, China
| | - Jieqiong Tan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
8
|
Abstract
Endometriosis is an inflammatory disease marked by ectopic growth of endometrial cells. Mesenchymal stromal cells (MSC) have immunosuppressive properties that have been suggested as a treatment for inflammatory diseases. Therefore, the aim herein was to examine effects of allogeneic MSC on endometriosis-derived cells in vitro as a potential therapy for endometriosis. MSC from allogeneic adipose tissue (Ad-MSC) and stromal cells from endometrium (ESCendo) and endometriotic ovarian cysts (ESCcyst) from women with endometriosis were isolated. The effects of Ad-MSC on ESCendo and ESCcyst were investigated using in vitro proliferation, apoptosis, adhesion, tube formation, migration, and invasion assays. Ad-MSC significantly increased proliferation of ESC compared to untreated controls. Moreover, Ad-MSC significantly decreased apoptosis and increased survival of ESC. Ad-MSC significantly increased adhesion of ESCendo and not ESCcyst on fibronectin. Conditioned medium from cocultures of Ad-MSC and ESC significantly increased tube formation of human umbilical vein endothelial cells on matrigel. Ad-MSC may significantly increase migration of ESCcyst and did not increase invasion of both cell types. The data suggest that allogeneic Ad-MSC should not be considered as a potential therapy for endometriosis, because they may support the pathology by maintaining and increasing growth of ectopic endometrial tissue.
Collapse
|
9
|
Yang HL, Mei J, Chang KK, Zhou WJ, Huang LQ, Li MQ. Autophagy in endometriosis. Am J Transl Res 2017; 9:4707-4725. [PMID: 29218074 PMCID: PMC5714760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 09/23/2017] [Indexed: 06/07/2023]
Abstract
Endometriosis (EMS) is a common gynecologic disease that causes chronic pelvic pain, dysmenorrhea, and infertility in women. The doctrine of menstruation back flow planting and defects in the immune system are well known and widely accepted. In recent years, increasing studies have been focused on the role of autophagy in EMS, and have shown that autophagy plays a vital role in EMS. Autophagy, which is known as the non-apoptotic form of programmed cell death induced by a large number of intracellular/extracellular stimuli, is the major cellular pathway for the degradation of long-lived proteins and cytoplasmic organelles in eukaryotic cells. Autophagy commonly refers to macroautophagy, which is characterized by autophagosomes (double-membrane vesicles). In normal endometrial tissues, autophagy is induced in glandular epithelial and stromal cells throughout the menstrual cycle. However, aberrant autophagy occurs in the eutopic endometrium and ectopic endometriotic foci, which contributes to the pathogenesis of EMS by promoting the hyperplasia of endometriotic tissues and stromal cells, restricting apoptosis, and inducing abnormal immune responses. Consistent with changes in autophagy levels between normal endometria, eutopic and ectopic endometria from patients with EMS, the altered expression of autophagy-related genes (ATGs) is also observed. Currently, many factors are involved in the aberrant autophagy of endometriotic tissues, including female hormones, certain drugs, hypoxia, and oxidative stress. Therefore, studies focusing on autophagy may uncover a new potential treatment for EMS. The aim of this review is to discuss the role of aberrant autophagy in EMS and to explore the potential value of autophagy as a target for EMS therapy.
Collapse
Affiliation(s)
- Hui-Li Yang
- Insititute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan UniversityShanghai 200011, People’s Republic of China
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan UniversityShanghai 200032, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, People’s Republic of China
| | - Jie Mei
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjing 210008, Jiangsu, People’s Republic of China
| | - Kai-Kai Chang
- Insititute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan UniversityShanghai 200011, People’s Republic of China
| | - Wen-Jie Zhou
- Insititute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan UniversityShanghai 200011, People’s Republic of China
| | - Li-Qing Huang
- Department of Statistics and Psychology, College of Letters and Science, University of California DavisDavis, CA 95618, USA
| | - Ming-Qing Li
- Insititute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan UniversityShanghai 200011, People’s Republic of China
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan UniversityShanghai 200032, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, People’s Republic of China
| |
Collapse
|
10
|
Zhan L, Li J, Wei B. Autophagy in endometriosis: Friend or foe? Biochem Biophys Res Commun 2017; 495:60-63. [PMID: 29107692 DOI: 10.1016/j.bbrc.2017.10.145] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 01/18/2023]
Abstract
Endometriosis is a chronic, estrogen-dependent disease and characterized by the implantation of endometrial glands and stroma deep and haphazardly into the outside the uterine cavity. It affects an estimated 10% of the female population of reproductive age and results in obvious reduction in health-related quality of life. Unfortunately, there is no a consistent theory for the etiology of endometriosis. Furthermore, the endometriosis is hard to diagnose in early stage and the treatment methods are limited. Importantly, emerging evidence has investigated that there is a close relationship between endometriosis and autophagy. However, autophagy is a friend or foe in endometriosis is puzzling, the precise mechanism underlying autophagy in endometriosis has not been fully elucidated yet. Here, we provide an integrated view on the acquired findings of the connections between endometriosis and autophagy. We also discuss which may contribute to the abnormal level of autophagy in endometriosis.
Collapse
Affiliation(s)
- Lei Zhan
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, 678 Fu Rong Road, Hefei 230601, China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Meishan Road No. 81, 230032 Hefei, China
| | - Bing Wei
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, 678 Fu Rong Road, Hefei 230601, China.
| |
Collapse
|
11
|
Kushnir VA, Seifer DB, Barad DH, Sen A, Gleicher N. Potential therapeutic applications of human anti-Müllerian hormone (AMH) analogues in reproductive medicine. J Assist Reprod Genet 2017; 34:1105-1113. [PMID: 28643088 PMCID: PMC5581791 DOI: 10.1007/s10815-017-0977-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/07/2017] [Indexed: 11/25/2022] Open
Abstract
Members of the transforming growth factor-beta (TGF-beta) superfamily are key regulators of various physiological processes. Anti-Müllerian hormone (AMH) which is also commonly known as Müllerian-inhibiting substance (MIS) is a member of the TGF-beta superfamily and an important regulator of reproductive organ differentiation and ovarian follicular development. While AMH has been used for diagnostic purposes as a biomarker for over 15 years, new potential therapeutic applications of recombinant human AMH analogues are now emerging as pharmacologic agents in reproductive medicine. Therapeutic uses of AMH in gonadal tissue may provide a unique opportunity to address a broad range of reproductive themes, like contraception, ovulation induction, onset of menopause, and fertility preservation, as well as specific disease conditions, such as polycystic ovarian syndrome (PCOS) and cancers of the reproductive tract. This review explores the most promising therapeutic applications for a novel class of drugs known as AMH analogues with agonist and antagonist functions.
Collapse
Affiliation(s)
- Vitaly A Kushnir
- Center for Human Reproduction, 21 East 69th Street, New York, NY, 10021, USA.
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - David B Seifer
- Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - David H Barad
- Center for Human Reproduction, 21 East 69th Street, New York, NY, 10021, USA
- Foundation for Reproductive Medicine, New York, NY, USA
| | - Aritro Sen
- Center for Human Reproduction, 21 East 69th Street, New York, NY, 10021, USA
- Division of Endocrinology & Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Norbert Gleicher
- Center for Human Reproduction, 21 East 69th Street, New York, NY, 10021, USA
- Foundation for Reproductive Medicine, New York, NY, USA
- Department of Obstetrics and Gynecology, University of Vienna School of Medicine, Vienna, Austria
- The Rockefeller University, New York, NY, USA
| |
Collapse
|
12
|
de Almeida Borges VR, Tavares MR, da Silva JH, Tajber L, Boylan F, Ribeiro AF, Nasciutti LE, Cabral LM, de Sousa VP. Development and characterization of poly(lactic-co-glycolic) acid nanoparticles loaded with copaiba oleoresin. Pharm Dev Technol 2017; 23:343-350. [DOI: 10.1080/10837450.2017.1290107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
| | - Marina R. Tavares
- Department of Pharmaceutics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julianna Henriques da Silva
- Research Program in Cellular Biology and Development, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lidia Tajber
- School of Pharmacy, Trinity College, University of Dublin, Dublin, Ireland
| | - Fabio Boylan
- School of Pharmacy, Trinity College, University of Dublin, Dublin, Ireland
| | - Ana Ferreira Ribeiro
- Department of Pharmaceutics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Eurico Nasciutti
- Research Program in Cellular Biology and Development, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucio Mendes Cabral
- Department of Pharmaceutics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
13
|
Park SH, Chung YJ, Song JY, Kim SI, Pépin D, MacLaughlin DT, Donahoe PK, Kim JH. Müllerian inhibiting substance inhibits an ovarian cancer cell line via β-catenin interacting protein deregulation of the Wnt signal pathway. Int J Oncol 2017; 50:1022-1028. [DOI: 10.3892/ijo.2017.3874] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/02/2017] [Indexed: 11/06/2022] Open
|
14
|
Ruiz LA, Báez-Vega PM, Ruiz A, Peterse DP, Monteiro JB, Bracero N, Beauchamp P, Fazleabas AT, Flores I. Dysregulation of Lysyl Oxidase Expression in Lesions and Endometrium of Women With Endometriosis. Reprod Sci 2015; 22:1496-508. [PMID: 25963914 PMCID: PMC5933196 DOI: 10.1177/1933719115585144] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Lysyl oxidases (LOXs) are enzymes involved in collagen deposition, extracellular membrane remodeling, and invasive/metastatic potential. Previous studies reveal an association of LOXs and endometriosis. We aimed to identify the mechanisms activated by upregulation of lysyl oxidases (LOX) in endometriotic cells and tissues. We hypothesized that LOX plays a role in endometriosis by promoting invasiveness and epithelial to mesenchymal transition (EMT). METHODS The LOX protein expression levels were measured by immunohistochemistry in lesions and endometrium on a tissue microarray (TMA) and in endometrial biopsies from patients and controls during the window of implantation (WOI). Estradiol regulation of LOX expression was determined by quantitative polymerase chain reaction (qPCR). Proliferation, invasion, and migration assays were performed in epithelial (endometrial epithelial cell), endometrial (human endometrial stromal cell), and endometriotic cell lines (ECL and 12Z). Pathway-focused multiplex qPCR was used to determine transcriptome changes due to LOX overexpression. RESULTS LOX protein was differentially expressed in ovarian versus peritoneal lesions. During WOI, LOX levels were higher in luminal epithelium of patients with endometriosis-associated infertility compared to controls. Invasive epithelial cell lines expressed higher levels of LOX than noninvasive ones. Transfection of LOX into noninvasive epithelial cells increased their migration in an LOX inhibitor-sensitive manner. Overexpression of LOX did not fully induce EMT but the expression of genes related to fibrosis and extracellular matrix remodeling were dysregulated. CONCLUSIONS This study documents that expression of LOX is differentially regulated in endometriotic lesions and endometrium. A role for LOX in mediating proliferation, migration, and invasion of endometrial and endometriotic cells was observed, which may be implicated in the establishment and progression of endometriotic lesions.
Collapse
Affiliation(s)
- Lynnette A Ruiz
- Department of Anatomy, Ponce Health Sciences University-School of Medicine & Ponce Research Institute, Ponce, PR, USA
| | - Perla M Báez-Vega
- Comprehensive Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, USA
| | - Abigail Ruiz
- Department of Microbiology, Ponce Health Sciences University-School of Medicine & Ponce Research Institute, Ponce, PR, USA
| | - Daniëlle P Peterse
- Department of Development and Regeneration, University Hospital Leuven, Leuven, Belgium
| | - Janice B Monteiro
- Department of Biochemistry, Ponce Health Sciences University- School of Medicine & Ponce Research Institute, Ponce, PR, USA
| | - Nabal Bracero
- Department of Ob-Gyn, University of Puerto Rico - Medical Sciences Campus, Genes Fertility Institute, San Juan, PR, USA
| | | | - Asgerally T Fazleabas
- Department of Ob-Gyn & Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Idhaliz Flores
- Department of Microbiology, Department of Ob-Gyn, Ponce Health Sciences University-School of Medicine & Ponce Research Institute, Ponce, PR, USA
| |
Collapse
|
15
|
Henriques da Silva J, Borges VRDA, Pereira LDCB, Ferrari R, de Mattos RM, Barros EGDO, Palmero CY, Fernandes PD, de Carvalho PR, Pereira de Sousa V, Cabral LM, Nasciutti LE. The oil-resin of the tropical rainforest tree Copaifera langsdorffii reduces cell viability, changes cell morphology and induces cell death in human endometriotic stromal cultures. ACTA ACUST UNITED AC 2015; 67:1744-55. [PMID: 26407531 DOI: 10.1111/jphp.12479] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 07/28/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The hormonal treatment for endometriosis frequently fails to completely eradicate endometriotic implants. A new therapeutic treatment is needed. This study investigates the in-vitro effect of Copaifera langsdorffii oil-resin on human eutopic and ectopic endometrium stromal cell cultures (EuESCs and EctESCs). METHODS A nanocomposite system containing the copaiba oil-resin (NanoCOR) was developed and acute toxicity test was performed. Endometrial stromal cells (ESCs) from non-endometriotics controls (CESCs), EuESCs and EctESCs were isolated and treated with different concentrations of NanoCOR, at different time intervals to evaluate its effect on cell morphology, proliferation, viability, necrosis and apoptosis induction. KEY FINDINGS When treated with 50 μg/ml of NanoCOR, the morphology of EctESCs changed, as the actin microfilaments were disorganized, disassembled or disrupted. Moreover, at 24 h of treatment with NanoCOR, the EctESCs viability was inhibited, and a significant number of these cells underwent apoptosis. In EuESCs, these effects were observed only at 48 h. Finally, the treatment of EctESCs with NanoCOR increased the lactate dehydrogenase release into the extracellular medium more than in EuESCs. CONCLUSIONS Our data indicate that NanoCOR has a greater impact on the behaviour of human endometriotic stromal cells than on the eutopic endometrium stromal cells, supporting the idea that NanoCOR should be further investigated as a novel and valuable alternative to treat endometriosis.
Collapse
Affiliation(s)
- Julianna Henriques da Silva
- Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Vinicius Raphael de Almeida Borges
- Laboratório de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Leonardo da Cunha Boldrini Pereira
- Laboratório de Bioengenharia Tecidual, Diretoria de Metrologia Aplicada às Ciências da Vida, Instituto Nacional de Metrologia, Qualidade e Tecnologia - Inmetro, Xerém, Rio de Janeiro, Brazil
| | - Renato Ferrari
- Instituto de Ginecologia da Universidade Federal do Rio de Janeiro, Hospital Universitário Moncorvo Filho, Centro, Rio de Janeiro, Brazil
| | - Rômulo Medina de Mattos
- Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Eliane Gouveia de Oliveira Barros
- Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Celia Yelimar Palmero
- Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Patricia Dias Fernandes
- Laboratório de Farmacologia da Inflamação e Óxido Nítrico, Programa de Farmacologia Celular e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Patricia Ribeiro de Carvalho
- Laboratório de Farmacologia da Inflamação e Óxido Nítrico, Programa de Farmacologia Celular e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Valeria Pereira de Sousa
- Laboratório de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Lucio Mendes Cabral
- Laboratório de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| | - Luiz Eurico Nasciutti
- Programa de Pesquisa em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão, Xerém, Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Müllerian inhibiting substance/anti-Müllerian hormone: A novel treatment for gynecologic tumors. Obstet Gynecol Sci 2014; 57:343-57. [PMID: 25264524 PMCID: PMC4175594 DOI: 10.5468/ogs.2014.57.5.343] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/15/2014] [Accepted: 05/15/2014] [Indexed: 01/02/2023] Open
Abstract
Müllerian inhibiting substance (MIS), also called anti-Müllerian hormone (AMH), is a member of the transforming growth factor-β super-family of growth and differentiation response modifiers. It is produced in immature Sertoli cells in male embryos and binds to MIS/AMH receptors in primordial Müllerian ducts to cause regression of female reproductive structures that are the precursors to the fallopian tubes, the surface epithelium of the ovaries, the uterus, the cervix, and the upper third of the vagina. Because most gynecologic tumors originate from Müllerian duct-derived tissues, and since MIS/AMH causes regression of the Müllerian duct in male embryos, it is expected to inhibit the growth of gynecologic tumors. Purified recombinant human MIS/AMH causes growth inhibition of epithelial ovarian cancer cells and cell lines in vitro and in vitro via MIS receptor-mediated mechanism. Furthermore, several lines of evidence suggest that MIS/AMH inhibits proliferation in tissues and cell lines of other MIS/AMH receptor-expressing gynecologic tumors such as cervical, endometrial, breast, and in endometriosis as well. These findings indicate that bioactive MIS/AMH recombinant protein should be tested in patients against tumors expressing the MIS/AMH receptor complex, perhaps beginning with ovarian cancer because it has the worst prognosis. The molecular tools to identify MIS/AMH receptor expressing ovarian and other cancers are in place, thus, it is possible to select patients for treatment. An MIS/AMH ELISA exists to follow administered doses of MIS/AMH, as well. Clinical trials await the production of sufficient supplies of qualified recombinant human MIS/AMH for this purpose.
Collapse
|
17
|
Signorile PG, Petraglia F, Baldi A. Anti-mullerian hormone is expressed by endometriosis tissues and induces cell cycle arrest and apoptosis in endometriosis cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:46. [PMID: 24886254 PMCID: PMC4046500 DOI: 10.1186/1756-9966-33-46] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 05/19/2014] [Indexed: 01/15/2023]
Abstract
Background The anti-mullerian hormone (AMH) is a member of the transforming growth factor β (TGF-β) superfamily, which is responsible of the regression of the mullerian duct. AMH is expressed in the normal endometrium, where, acting in a paracrine fashion, negatively regulates cellular viability. Our objective was to evaluate the in vitro effects of the treatment with AMH of endometriosic cells. Methods AMH expression in human endometriosis glands was evaluated by immunohistochemistry. RT-PCR has been used to quantify the expression levels of AMH and AMH RII isoforms, as well as of cytochrome P450 in both endometriosis epithelial and stromal cells Effects of AMH and AMH-cleaved treatment in endometriosis cells were evaluated by flow-cytometry analysis. Finally, it has been evaluated the effect of plasmin-digested AMH on cytochrome P450 activity. Results AMH and AMH RII isoforms, as well as cytochrome P450, were expressed in both endometriosis epithelial and stromal cells. Treatment of endometriosis stromal and epithelial cell growth with AMH was able to induce a decrease in the percentage of cells in S phase and increase percentage of cells in G1 and G2 phase; coherently, decreased cell viability and increased percentage of cells death fraction was observed. The plasmin-digested AMH was able to suppress most of the cytochrome P450 activity, causing an increase of pre-G1 phase and of apoptosis induction treating with plasmin-digested AMH in both cell lines, most marked in the epithelial cells. Conclusions The data produced suggest a possible use of AMH as therapeutic agents in endometriosis.
Collapse
|
18
|
Hemming S, Cakouros D, Gronthos S. Detachment of mesenchymal stem cells with trypsin/EDTA has no effect on apoptosis detection. Stem Cells 2014; 32:1991-2. [PMID: 24677681 DOI: 10.1002/stem.1710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 03/17/2014] [Indexed: 11/12/2022]
Affiliation(s)
- Sarah Hemming
- Mesechymal Stem Cell Laboratory, School of Health Sciences, Faculty of Health Sciences; Centre for Stem Cell Research, University of Adelaide, South Australia, Australia
| | | | | |
Collapse
|