1
|
Shi L, Ying H, Dai Y, Rong Y, Chen J, Zhou F, Wang S, Xu S, Tong X, Zhang S. Upregulated let-7 expression in the follicular fluid of patients with endometriomas leads to dysfunction of granulosa cells through targeting of IGF1R. Hum Reprod 2025; 40:119-137. [PMID: 39521729 DOI: 10.1093/humrep/deae247] [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: 05/28/2024] [Revised: 09/07/2024] [Indexed: 11/16/2024] Open
Abstract
STUDY QUESTION What molecular mechanisms underlie the decline in ovarian reserve as the number and quality of oocytes decrease in patients with ovarian endometriomas (OEM)? SUMMARY ANSWER Elevated expression of the let-7 micro(mi)RNAs in the follicular microenvironment of OEM-affected ovaries targets the expression of type 1 insulin-like growth factor receptor (IGF1R) in granulosa cell (GC) and disrupts their proliferation, steroid hormone secretion levels, adenosine triphosphate (ATP) energy metabolism, and reactive oxygen species (ROS) oxidative stress levels. WHAT IS KNOWN ALREADY Patients with OEM exhibit diminished ovarian reserve, characterized by reduced oocyte quantity and quality. Fibrotic changes in the ovarian tissue surrounding the OEM create a disruptive microenvironment for follicular growth and development. STUDY DESIGN, SIZE, DURATION This is a cross-sectional study aimed to elucidate the molecular mechanisms underlying the impact of OEM on follicular development. Initially, miRNA expression profiles in follicular fluid (FF) samples were sequenced from patients with infertility related to OEM (N = 3) and male factor (MF) infertility (N = 3), with the latter serving as the control group. Differentially expressed miRNAs were validated in additional samples from each group (N = 55 in OEM group and N = 45 in MF group) to confirm candidate miRNAs. The study also investigated indicators associated with GCs dysfunction in vitro on rat GCs. Subsequently, rat models of OEM were established through endometrial allogeneic transplantation, and fertility experiments were conducted to assess the let-7/IGF1R axis response to OEM in vivo. Patient samples were collected between May 2018 and April 2019, and the mechanistic study was conducted over the subsequent three years. PARTICIPANTS/MATERIALS, SETTING, METHODS FF and GC samples were obtained from infertile patients undergoing IVF treatment for OEM and MF related infertility. miRNA expression profiles in FF samples were analyzed using second-generation high-throughput sequencing technology, and candidate miRNAs were validated through quantitative PCR (qPCR). In the in vitro experiments conducted with rat GCs, cell proliferation was assessed using the CCK-8 assay, while steroid hormone concentrations were measured using chemiluminescence. ATP content was determined with an ATP assay kit, and levels of ROS were quantified using flow cytometry. A dual luciferase reporter gene assay was employed to identify the target gene of let-7 based on the construction of a IGF1R reporter gene plasmid using 293T cells. Western blotting was utilized to evaluate the expression of IGF1R in GCs, as well as its downstream proteins, and changes in signaling pathways following let-7 agomir/antagomir transfection and/or Igf1r silencing. In the in vivo OEM rat models, alterations in ovarian structure and cyst morphology were observed using hematoxylin and eosin staining. The expressions of let-7 and Igf1r in GCs were evaluated through qPCR, while variations in IGF1R expression were investigated with immunohistochemistry. MAIN RESULTS AND THE ROLE OF CHANCE The cohort of patients with ovarian OEM in this study exhibited significantly decreased antral follicle counts, oocyte retrieval numbers, and normal fertilization rates compared to the control group with MF. The expression of the let-7 miRNA family was markedly upregulated in the FF and GCs of OEM patients. Transfection of rat GCs with let-7 agonists diminished the functions of GCs, including disrupted cell proliferation, mitochondrial oxidative phosphorylation, and steroid hormone secretion, while transfection of rat GCs with let-7 antagonists caused the opposite effects. Luciferase reporter gene experiments confirmed that let-7 complementarily bound to the 3'-untranslated regions of IGF1R. Stimulation of let-7 expression in rat GCs led to a significant decrease in IGF1R expression, while inhibition of let-7 increased IGF1R expression. The expression of IGF1R in the GCs of OEM patients was also significantly reduced compared to MF patients. Silencing of Igf1r led to the dysfunction of GCs, similar to the effects of let-7 agonization, as demonstrated by the downregulation of key proteins involved in cell proliferation (CCND2 and CCND3) and oestradiol synthesis, as well as an increase in progesterone synthesis (StAR), while implicating the PI3K-Akt and MAPK signaling pathways. The antagonistic effect of let-7 on GCs was ineffective when Igf1r was silenced. Conversely, the agonistic effect of let-7 on GCs could be reversed by stimulation with the IGF1R ligand IGF-1. These findings suggested that let-7 regulated the proliferation, differentiation, and ATP synthesis of GCs through targeting IGF1R. The OEM rat model demonstrated alterations in ovarian morphology and structure, along with reduced fertility. Let-7 expression was significantly upregulated in GCs of OEM rats compared to normal rats, while Igf1r and IGF1R expression in pre-ovulatory follicular GCs were notably downregulated, supporting the notion that elevated let-7 expression in the follicular microenvironment of OEM inhibited IGF1R, leading to abnormal GC function and impacting fertility at the molecular level. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The synthesis and secretion mechanisms of steroid hormones are intricate and complex. Some enzymes that regulate oestrogen synthesis also play a role in progesterone synthesis. Moreover, certain receptors can respond to multiple hormone signals. Therefore, in this study, the expression patterns of key enzymes such as CYP17A, CYP11A1, HSD3B2, StAR, and receptors including AR, LHCGR, FSHR, ESR2, might be influenced by various factors and might not demonstrate complete consistency. WIDER IMPLICATIONS OF THE FINDINGS Future research will concentrate on investigating the potential impact of ovarian stromal cells on the external microenvironment of follicle growth. Additionally, screening for small molecule drugs that target let-7 and IGF1R actions can be conducted to intervene and modify the ovarian microenvironment, ultimately enhancing ovarian function. STUDY FUNDING/COMPETING INTEREST(S) This study received funding from the National Natural Science Foundation of China (grant number 82301851 to L.B.S., grant numbers U23A20403 and U20A20349 to S.Y.Z., and grant number 82371637 to Y.D.D.) and the Natural Science Foundation of Zhejiang Province (grant LTGY23H040010 to F.Z.). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Libing Shi
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Hanqi Ying
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Yongdong Dai
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Yan Rong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Jianmin Chen
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Feng Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Shasha Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Shiqian Xu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Xiaomei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, PR China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, PR China
| |
Collapse
|
2
|
Rivera-Arce LA, Cruz ML, Rodriguez-Cintron U, Torres-Pirela JP, Appleyard CB. Implication of the enteric glia in the IBS-like colonic inflammation associated with endometriosis. BMC Womens Health 2024; 24:647. [PMID: 39707348 PMCID: PMC11662455 DOI: 10.1186/s12905-024-03480-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Endometriosis is a complex gynecological disorder characterized by the ectopic growth of endometrial tissue. Symptoms of endometriosis are known to impair the quality of life of patients, and among these are found dysmenorrhea, chronic pelvic pain, and gastrointestinal (GI) issues. GI issues such as painful bowel movements, bloating and constipation or diarrhea, are one of the common reasons for misdiagnosis with irritable bowel syndrome (IBS). Enteric glial cells (EGC) are known to play a role in pain associated with IBS, and reactive gliosis has been reported in patients with IBS, but the role of EGC in endometriosis has yet to be elucidated. We hypothesized that endometriosis will induce reactive gliosis, with increased expression of the glial fibrillary acidic protein (GFAP) and S100B, in the myenteric plexus of colonic sections in an animal model of endometriosis. METHODS In the present study animal experiments were employed to explore the impact of endometriosis on the gastrointestinal tract. Using a surgically induced endometriosis rat model, we collected ileal and colonic segments for analysis. We used H&E to assess microscopic damage in colon and ileum, immunofluorescence to measure GFAP and S100B expression in the colon, and toluidine blue staining to measure mast cell infiltration in colon and ileum. Immunofluorescence images were captured using confocal microscope and analyzed using ImageJ software. RESULTS All endometriosis animals developed vesicles. These animals had a significant increase in the colonic macroscopic damage compared to Sham and Naïve controls. Colonic and ileal sections didn't show statistical differences in microscopic damage between groups, yet endometriosis ileum had significantly increased mast cell infiltration compared to Naïve. GFAP immunostaining showed significantly increased integrated density in endometriosis when compared to Sham or Naïve, while no statistical difference was found in S100B integrated density between groups. CONCLUSIONS We conclude that endometriosis can alter GI homeostasis by inducing colon inflammation, reactive gliosis, and ileal mast cell infiltration. Taken together this suggests endometriosis can mimic IBS histopathology beyond the symptomatology, reinforcing this disease's complexity and the need to treat it beyond the gynecological setting.
Collapse
Affiliation(s)
- Luis A Rivera-Arce
- Department of Basic Sciences - Physiology Division, Ponce Health Sciences University, Ponce Research Institute, PO Box 7004, Ponce, 00732-7004, PR, Puerto Rico
| | - Myrella L Cruz
- Department of Basic Sciences - Physiology Division, Ponce Health Sciences University, Ponce Research Institute, PO Box 7004, Ponce, 00732-7004, PR, Puerto Rico
| | - Ulises Rodriguez-Cintron
- Department of Basic Sciences - Physiology Division, Ponce Health Sciences University, Ponce Research Institute, PO Box 7004, Ponce, 00732-7004, PR, Puerto Rico
| | - James P Torres-Pirela
- Department of Basic Sciences - Physiology Division, Ponce Health Sciences University, Ponce Research Institute, PO Box 7004, Ponce, 00732-7004, PR, Puerto Rico
| | - Caroline B Appleyard
- Department of Basic Sciences - Physiology Division, Ponce Health Sciences University, Ponce Research Institute, PO Box 7004, Ponce, 00732-7004, PR, Puerto Rico.
| |
Collapse
|
3
|
Anchan MM, Kalthur G, Datta R, Majumdar K, P K, Dutta R. Unveiling the fibrotic puzzle of endometriosis: An overlooked concern calling for prompt action. F1000Res 2024; 13:721. [PMID: 39669683 PMCID: PMC11635194 DOI: 10.12688/f1000research.152368.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/14/2024] Open
Abstract
Endometriosis is a benign, estrogen-dependent, persistent chronic inflammatory heterogeneous condition that features fibrotic adhesions caused by periodic bleeding. The characteristic ectopic lesions are marked by a widely spread dense fibrotic interstitium comprising of fibroblasts, myofibroblasts, collagen fibers, extracellular proteins, inflammatory cells, and active angiogenesis. Fibrosis is now recognized as a critical component of endometriosis because of which current treatments, such as hormonal therapy and surgical excision of lesions are largely ineffective with severe side effects, high recurrence rates, and significant morbidity. The symptoms include dysmenorrhea (cyclic or noncyclic), dyspareunia, abdominal discomfort, and infertility. The significant lack of knowledge regarding the underlying root causes, etiology, and complex pathogenesis of this debilitating condition, hinders early diagnosis and implement effective therapeutic approaches with minimal side effects presenting substantial hurdles in endometriosis management. Emerging research offer a close relationship between endometriosis and fibrosis, which is believed to be tightly linked to pain, a primary contributor to the deterioration of the patient's quality of life. However, the underlying pathophysiological cellular and molecular signaling pathways behind endometriosis-associated fibrosis are poorly addressed. The available experimental disease models have tremendous challenges in reproducing the human characteristics of the disease limiting the treatment effectiveness. Future translational research on the topic has been hindered by the lack of an adequate fibrotic model of endometriosis emphasizing the necessity of etiological exploration. This review article focuses on recent developments in the field and highlight the necessity for novel fibrotic models for early diagnosis, a better understanding the disease's etiology and develop effective anti-fibrotic treatments. By addressing these knowledge gaps, we want to open fresh avenues for a thorough investigation and extended research in the field of endometriosis.
Collapse
Affiliation(s)
- Megha M Anchan
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Guruprasad Kalthur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | | | - Kabita Majumdar
- Gauhati Medical College & Hospital IVF centre, Bhangagarh, Gauhati Medical College, Assam, 781032, India
| | - Karthikeyan P
- Department of General Surgery, Government Kallakurichi Medical College, Government Kallakurichi Medical College, Kallakurichi, Tamil Nadu, India
| | - Rahul Dutta
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
4
|
Panir K, Schjenken JE, Breen J, Chan HY, Greaves E, Robertson SA, Hull ML. RNA sequencing reveals molecular mechanisms of endometriosis lesion development in mice. Dis Model Mech 2024; 17:dmm050566. [PMID: 39385609 PMCID: PMC11524442 DOI: 10.1242/dmm.050566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
Understanding of molecular mechanisms contributing to the pathophysiology of endometriosis, and upstream drivers of lesion formation, remains limited. Using a C57Bl/6 mouse model in which decidualized endometrial tissue is injected subcutaneously in the abdomen of recipient mice, we generated a comprehensive profile of gene expression in decidualized endometrial tissue (n=4), and in endometriosis-like lesions at Day 7 (n=4) and Day 14 (n=4) of formation. High-throughput mRNA sequencing allowed identification of genes and pathways involved in the initiation and progression of endometriosis-like lesions. We observed distinct patterns of gene expression with substantial differences between the lesions and the decidualized endometrium that remained stable across the two lesion timepoints, and showed similarity to transcriptional changes implicated in human endometriosis lesion formation. Pathway enrichment analysis revealed several immune and inflammatory response-associated canonical pathways, multiple potential upstream regulators, and involvement of genes not previously implicated in endometriosis pathogenesis, including IRF2BP2 and ZBTB10, suggesting novel roles in disease progression. Collectively, the provided data will be a useful resource to inform research on the molecular mechanisms contributing to endometriosis-like lesion development in this mouse model.
Collapse
Affiliation(s)
- Kavita Panir
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - John E. Schjenken
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - James Breen
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- South Australian Genomics Centre (SAGC), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Computational and Systems Biology Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | - Hon Yeung Chan
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Erin Greaves
- Centre for Early Life, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Sarah A. Robertson
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - M. Louise Hull
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Department of Obstetrics and Gynaecology, Women's and Children's Hospital, Adelaide, SA 5006, Australia
| |
Collapse
|
5
|
Crawford AJ, Forjaz A, Bons J, Bhorkar I, Roy T, Schell D, Queiroga V, Ren K, Kramer D, Huang W, Russo GC, Lee MH, Wu PH, Shih IM, Wang TL, Atkinson MA, Schilling B, Kiemen AL, Wirtz D. Combined assembloid modeling and 3D whole-organ mapping captures the microanatomy and function of the human fallopian tube. SCIENCE ADVANCES 2024; 10:eadp6285. [PMID: 39331707 PMCID: PMC11430475 DOI: 10.1126/sciadv.adp6285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/22/2024] [Indexed: 09/29/2024]
Abstract
The fallopian tubes play key roles in processes from pregnancy to ovarian cancer where three-dimensional (3D) cellular and extracellular interactions are important to their pathophysiology. Here, we develop a 3D multicompartment assembloid model of the fallopian tube that molecularly, functionally, and architecturally resembles the organ. Global label-free proteomics, innovative assays capturing physiological functions of the fallopian tube (i.e., oocyte transport), and whole-organ single-cell resolution mapping are used to validate these assembloids through a multifaceted platform with direct comparisons to fallopian tube tissue. These techniques converge at a unique combination of assembloid parameters with the highest similarity to the reference fallopian tube. This work establishes (i) an optimized model of the human fallopian tubes for in vitro studies of their pathophysiology and (ii) an iterative platform for customized 3D in vitro models of human organs that are molecularly, functionally, and microanatomically accurate by combining tunable assembloid and tissue mapping methods.
Collapse
Affiliation(s)
- Ashleigh J Crawford
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - André Forjaz
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Isha Bhorkar
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Triya Roy
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - David Schell
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Vasco Queiroga
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kehan Ren
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Donald Kramer
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biotechnology, Johns Hopkins Advanced Academic Programs, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wilson Huang
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gabriella C Russo
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Meng-Horng Lee
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Pei-Hsun Wu
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ie-Ming Shih
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tian-Li Wang
- Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark A Atkinson
- Departments of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
- Departments of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | | | - Ashley L Kiemen
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Functional Anatomy and Evolution, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Denis Wirtz
- Johns Hopkins Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Marchandot B, Faller E, Akladios C, Matsushita K, Bäck M, Jesel L, Schini-Kerth V, Morel O. Fostering cardio-endometriosis: a call to action for a comprehensive understanding of cardiovascular disease in endometriosis. Eur J Prev Cardiol 2024; 31:1574-1582. [PMID: 38421615 DOI: 10.1093/eurjpc/zwae087] [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: 12/14/2023] [Revised: 02/06/2024] [Accepted: 02/17/2024] [Indexed: 03/02/2024]
Abstract
Recently, a growing body of evidence has highlighted a concerning link between endometriosis and cardiovascular disease. Endometriosis, a chronic, inflammatory, hormone-dependent condition affecting 5-10% of reproductive-aged women worldwide, has long been associated with reproductive and gynaecological consequences. However, emerging research has suggested that it may also contribute to adverse cardiovascular outcomes. This paper aims to shed light on the importance of recognizing cardio-endometriosis as a new and developing sphere of research in the field of cardiology, thereby urging the medical community to address this pressing issue.
Collapse
Affiliation(s)
- Benjamin Marchandot
- Division of Cardiovascular Medicine, Strasbourg University Hospital, 1 Place de l'Hopital, 67000 Strasbourg, France
- UR 3074 Médecine Cardiovasculaire Translationnelle, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France
| | - Emilie Faller
- Department of Obstetrics and Gynecology, Hautepierre Hospital, University Hospital, 1 Avenue Molière, 67000 Strasbourg, France
- ENDOALSACE, Strasbourg Expert Center for Endometriosis, Hautepierre Hospital, University Hospital, 1 Avenue Molière, 67000 Strasbourg, France
| | - Cherif Akladios
- Department of Obstetrics and Gynecology, Hautepierre Hospital, University Hospital, 1 Avenue Molière, 67000 Strasbourg, France
- ENDOALSACE, Strasbourg Expert Center for Endometriosis, Hautepierre Hospital, University Hospital, 1 Avenue Molière, 67000 Strasbourg, France
| | - Kensuke Matsushita
- UR 3074 Médecine Cardiovasculaire Translationnelle, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Magnus Bäck
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
- Section of Translational Cardiology, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden
- Université de Lorraine, Institut National de la Sante et de la Recherche Medicale U1116, Nancy, France
| | - Laurence Jesel
- Division of Cardiovascular Medicine, Strasbourg University Hospital, 1 Place de l'Hopital, 67000 Strasbourg, France
- UR 3074 Médecine Cardiovasculaire Translationnelle, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France
| | - Valérie Schini-Kerth
- UR 3074 Médecine Cardiovasculaire Translationnelle, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France
| | - Olivier Morel
- Division of Cardiovascular Medicine, Strasbourg University Hospital, 1 Place de l'Hopital, 67000 Strasbourg, France
- UR 3074 Médecine Cardiovasculaire Translationnelle, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France
- Hanoï Medical University, Vietnam
| |
Collapse
|
7
|
Davis EHS, Jones C, Coward K. Rethinking the application of nanoparticles in women's reproductive health and assisted reproduction. Nanomedicine (Lond) 2024; 19:1231-1251. [PMID: 38686941 PMCID: PMC11285225 DOI: 10.2217/nnm-2023-0346] [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: 11/29/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
Nanoparticles and nanotechnology may present opportunities to revolutionize the prevention, treatment and diagnosis of a range of reproductive health conditions in women. These technologies are also used to improve outcomes of assisted reproductive technology. We highlight a range of these potential clinical uses of nanoparticles for polycystic ovary syndrome, endometriosis, uterine fibroids and sexually transmitted infections, considering in vitro and in vivo studies along with clinical trials. In addition, we discuss applications of nanoparticles in assisted reproductive technology, including sperm loading, gamete and embryo preservation and preventing preterm birth. Finally, we present some of the concerns associated with the medical use of nanoparticles, identifying routes for further exploration before nanoparticles can be applied to women's reproductive health in the clinic.
Collapse
Affiliation(s)
- Emily HS Davis
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Celine Jones
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Kevin Coward
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
8
|
Khan KN, de Ziegler D, Guo SW. Bacterial infection in endometriosis: a silver-lining for the development of new non-hormonal therapy? Hum Reprod 2024; 39:623-631. [PMID: 38300227 DOI: 10.1093/humrep/deae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/24/2023] [Indexed: 02/02/2024] Open
Abstract
The pathogenesis of endometriosis is a hotly debated topic, yet still cloaked in multiple layers of hypothetical theories. A recent report raises the possibility that bacterial infection, especially those of the genus Fusobacterium, may be the cause of endometriosis, at least in certain women. More importantly, the demonstration that treatment with broad-spectrum antibiotics significantly reduced the size of lesions in a mouse endometriosis model rekindles the hope for new non-hormonal treatments. The development of new therapies has been plagued by strings of unsuccessful clinical trials over the last two decades. Is this antibiotic therapy, a silver lining for the research and development of non-hormonal drugs for endometriosis?
Collapse
Affiliation(s)
- Khaleque N Khan
- Department of Obstetrics and Gynecology, The Clinical and Translational Research Center, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Sun-Wei Guo
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
- Research Institute, Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Lv Q, Zhang Y, Yang R, Dai Y, Lin Y, Sun K, Xu H, Tao K. Photoacoustic Imaging Endometriosis Lesions with Nanoparticulate Polydopamine as a Contrast Agent. Adv Healthc Mater 2024; 13:e2302175. [PMID: 37742067 DOI: 10.1002/adhm.202302175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/21/2023] [Indexed: 09/25/2023]
Abstract
Endometriosis (EM) is a prevalent and debilitating gynecological disorder primarily affecting women of reproductive age. The diagnosis of EM is historically hampered by delays, owing to the absence of reliable diagnostic and monitoring techniques. Herein, it is reported that photoacoustic imaging can be a noninvasive modality for deep-seated EM by employing a hyaluronic-acid-modified polydopamine (PDA@HA) nanoparticle as the contrast agent. The PDA@HA nanoparticles exhibit inherent absorption and photothermal effects when exposed to near-infrared light, proficiently converting thermal energy into sound waves. Leveraging the targeting properties of HA, distinct photoacoustic signals emanating from the periphery of orthotopic EM lesions are observed. These findings are corroborated through anatomical observations and in vivo experiments involving mice with green fluorescent protein-labeled EM lesions. Moreover, the changes in photoacoustic intensity over a 24 h period reflect the dynamic evolution of PDA@HA nanoparticle biodistribution. Through the utilization of a photoacoustic ultrasound modality, in vivo assessments of EM lesion volumes are conducted. This innovative approach not only facilitates real-time monitoring of the therapeutic kinetics of candidate drugs but also obviates the need for the sacrifice of experimental mice. As such, this study presents a promising avenue for enhancing the diagnosis and drug-screening processes of EM.
Collapse
Affiliation(s)
- Quanjie Lv
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yili Zhang
- International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Ruihao Yang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yingfan Dai
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yu Lin
- International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Kang Sun
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Hong Xu
- International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Ke Tao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
10
|
Slayden OD, Luo F, Martin D V M LD. A protocol for creating endometriosis in rhesus macaques (Macaca mulatta). J Med Primatol 2023; 52:405-413. [PMID: 37849073 PMCID: PMC10843508 DOI: 10.1111/jmp.12681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Endometriosis is the presence of endometrium-like tissue outside the uterine cavity. An experimental model of endometriosis has been created in the baboon by the transcervical collection and laparoscopic inoculation of menstrual endometrium. Macaques are the preferred model for pharmaceutical development, but the complex anatomy of the macaque cervix makes the baboon method impractical. In this work, we sought to validate a surgical approach for creating endometriosis in macaques. METHODS Menstrual endometrium was collected via laparoscopic intrauterine puncture and transferred to the peritoneal cavity. We repeated this procedure during three menstruations. Endometriotic tissue was identified during laparoscopy, collected, and characterized by immunohistochemistry. RESULTS Sham surgery-treated animals (n = 3) failed to develop endometriosis. We identified red, powder burnt, and white lesions in 13/14 of the treated animals; the stroma of the red lesions stained positive for ovarian steroid receptors. CONCLUSION This surgical technique can reliably create hormone-responsive endometriosis in macaques for therapeutic studies.
Collapse
Affiliation(s)
- Ov D Slayden
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon, USA
| | - Fangzhou Luo
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Lauren D Martin D V M
- Division of Animal Resources & Research Support, Oregon National Primate Research Center, Beaverton, Oregon, USA
| |
Collapse
|
11
|
Tan Z, Gong X, Wang CC, Zhang T, Huang J. Diminished Ovarian Reserve in Endometriosis: Insights from In Vitro, In Vivo, and Human Studies-A Systematic Review. Int J Mol Sci 2023; 24:15967. [PMID: 37958954 PMCID: PMC10647261 DOI: 10.3390/ijms242115967] [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: 08/31/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Endometriosis, a prevalent disorder in women of reproductive age, is often associated with undesired infertility. Ovarian reserve, an essential measure of ovarian function that is crucial for maintaining fecundity, is frequently diminished in women with endometriosis. Though the causative relationship between endometriosis and reduced ovarian reserve is not fully understood due to the lack of standardized and precise measurements of ovarian reserve, there is ongoing discussion regarding the impact of interventions for endometriosis on ovarian reserve. Therefore, in this review, we investigate articles that have related keywords and which were also published in recent years. Thereafter, we provide a comprehensive summary of evidence from in vitro, in vivo, and human studies, thereby shedding light on the decreased ovarian reserve in endometriosis. This research consolidates evidence from in vitro, in vivo, and human studies on the diminished ovarian reserve associated with endometriosis, as well as enhances our understanding of whether and how endometriosis, as well as its interventions, contribute to reductions in ovarian reserve. Furthermore, we explore potential strategies to modify existing therapy options that could help prevent diminished ovarian reserve in patients with endometriosis.
Collapse
Affiliation(s)
- Zhouyurong Tan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (Z.T.); (X.G.); (C.C.W.)
| | - Xue Gong
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (Z.T.); (X.G.); (C.C.W.)
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (Z.T.); (X.G.); (C.C.W.)
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Reproduction and Development, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tao Zhang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (Z.T.); (X.G.); (C.C.W.)
| | - Jin Huang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (Z.T.); (X.G.); (C.C.W.)
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
12
|
Al Thani NA, Hasan M, Yalcin HC. Use of Animal Models for Investigating Cardioprotective Roles of SGLT2 Inhibitors. J Cardiovasc Transl Res 2023; 16:975-986. [PMID: 37052784 PMCID: PMC10615955 DOI: 10.1007/s12265-023-10379-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023]
Abstract
Sodium-glucose co-transporter 2 (SGLT2) inhibitors represent one type of new-generation type 2 diabetes (T2DM) drug treatment. The mechanism of action of an SGLT2 inhibitor (SGLT2i) in treating T2DM depends on lowering blood glucose levels effectively via increasing the glomerular excretion of glucose. A good number of randomized clinical trials revealed that SGLT2is significantly prevented heart failure (HF) and cardiovascular death in T2DM patients. Despite ongoing clinical trials in HF patients without T2DM, there have been a limited number of translational studies on the cardioprotective properties of SGLT2is. As the cellular mechanism behind the cardiac benefits of SGLT2is is still to be elucidated, animal models are used to better understand the pathways behind the cardioprotective mechanism of SGLT2i. In this review, we summarize the animal models constructed to study the cardioprotective mechanisms of SGLT2is to help deliver a more comprehensive understanding of the in vivo work that has been done in this field and to help select the most optimal animal model to use when studying the different cardioprotective effects of SGLT2is.
Collapse
Affiliation(s)
- Najlaa A Al Thani
- Research and Development Department, Barzan Holdings, P. O. Box 7178, Doha, Qatar
| | - Maram Hasan
- Biomedical Research Center, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Huseyin C Yalcin
- Biomedical Research Center, Qatar University, P. O. Box 2713, Doha, Qatar.
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar.
| |
Collapse
|
13
|
Talebloo N, Bernal MAO, Kenyon E, Mallett CL, Fazleabas A, Moore A. Detection of Endometriosis Lesions Using Gd-Based Collagen I Targeting Probe in Murine Models of Endometriosis. Mol Imaging Biol 2023; 25:833-843. [PMID: 37418136 PMCID: PMC10598151 DOI: 10.1007/s11307-023-01833-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 07/08/2023]
Abstract
PURPOSE Endometriosis is a chronic condition characterized by high fibrotic content and affecting about 10% of women during their reproductive years. Yet, no clinically approved agents are available for non-invasive endometriosis detection. The purpose of this study was to investigate the utility of a gadolinium-based collagen type I targeting probe (EP-3533) to non-invasively detect endometriotic lesions using magnetic resonance imaging (MRI). Previously, this probe has been used for detection and staging of fibrotic lesions in the liver, lung, heart, and cancer. In this study we evaluate the potential of EP-3533 for detecting endometriosis in two murine models and compare it with a non-binding isomer (EP-3612). PROCEDURES For imaging, we utilized two GFP-expressing murine models of endometriosis (suture model and injection model) injected intravenously with EP3533 or EP-33612. Mice were imaged before and after bolus injection of the probes. The dynamic signal enhancement of MR T1 FLASH images was analyzed, normalized, and quantified, and the relative location of lesions was validated through ex vivo fluorescence imaging. Subsequently, the harvested lesions were stained for collagen, and their gadolinium content was quantified by inductively coupled plasma optical emission spectrometry (ICP-OES). RESULTS We showed that EP-3533 probe increased the signal intensity in T1-weighted images of endometriotic lesions in both models of endometriosis. Such enhancement was not detected in the muscles of the same groups or in endometriotic lesions of mice injected with EP-3612 probe. Consequentially, control tissues had significantly lower gadolinium content, compared to the lesions in experimental groups. Probe accumulation was similar in endometriotic lesions of either model. CONCLUSIONS This study provides evidence for feasibility of targeting collagen type I in the endometriotic lesions using EP3533 probe. Our future work includes investigation of the utility of this probe for therapeutic delivery in endometriosis to inhibit signaling pathways that cause the disease.
Collapse
Affiliation(s)
- Nazanin Talebloo
- Precision Health Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
| | - Maria Ariadna Ochoa Bernal
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI, 49503, USA
- Department of Animal Science, Michigan State University, 474 S Shaw Ln, East Lansing, MI, 48824, USA
| | - Elizabeth Kenyon
- Precision Health Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Christiane L Mallett
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, MI, 48824, USA
| | - Asgerally Fazleabas
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI, 49503, USA
| | - Anna Moore
- Precision Health Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA.
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
14
|
Szukiewicz D. Insight into the Potential Mechanisms of Endocrine Disruption by Dietary Phytoestrogens in the Context of the Etiopathogenesis of Endometriosis. Int J Mol Sci 2023; 24:12195. [PMID: 37569571 PMCID: PMC10418522 DOI: 10.3390/ijms241512195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Phytoestrogens (PEs) are estrogen-like nonsteroidal compounds derived from plants (e.g., nuts, seeds, fruits, and vegetables) and fungi that are structurally similar to 17β-estradiol. PEs bind to all types of estrogen receptors, including ERα and ERβ receptors, nuclear receptors, and a membrane-bound estrogen receptor known as the G protein-coupled estrogen receptor (GPER). As endocrine-disrupting chemicals (EDCs) with pro- or antiestrogenic properties, PEs can potentially disrupt the hormonal regulation of homeostasis, resulting in developmental and reproductive abnormalities. However, a lack of PEs in the diet does not result in the development of deficiency symptoms. To properly assess the benefits and risks associated with the use of a PE-rich diet, it is necessary to distinguish between endocrine disruption (endocrine-mediated adverse effects) and nonspecific effects on the endocrine system. Endometriosis is an estrogen-dependent disease of unknown etiopathogenesis, in which tissue similar to the lining of the uterus (the endometrium) grows outside of the uterus with subsequent complications being manifested as a result of local inflammatory reactions. Endometriosis affects 10-15% of women of reproductive age and is associated with chronic pelvic pain, dysmenorrhea, dyspareunia, and infertility. In this review, the endocrine-disruptive actions of PEs are reviewed in the context of endometriosis to determine whether a PE-rich diet has a positive or negative effect on the risk and course of endometriosis.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
15
|
Crawford AJ, Forjaz A, Bhorkar I, Roy T, Schell D, Queiroga V, Ren K, Kramer D, Bons J, Huang W, Russo GC, Lee MH, Schilling B, Wu PH, Shih IM, Wang TL, Kiemen A, Wirtz D. Precision-engineered biomimetics: the human fallopian tube. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543923. [PMID: 37333379 PMCID: PMC10274705 DOI: 10.1101/2023.06.06.543923] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The fallopian tube has an essential role in several physiological and pathological processes from pregnancy to ovarian cancer. However, there are no biologically relevant models to study its pathophysiology. The state-of-the-art organoid model has been compared to two-dimensional tissue sections and molecularly assessed providing only cursory analyses of the model's accuracy. We developed a novel multi-compartment organoid model of the human fallopian tube that was meticulously tuned to reflect the compartmentalization and heterogeneity of the tissue's composition. We validated this organoid's molecular expression patterns, cilia-driven transport function, and structural accuracy through a highly iterative platform wherein organoids are compared to a three-dimensional, single-cell resolution reference map of a healthy, transplantation-quality human fallopian tube. This organoid model was precision-engineered to match the human microanatomy. One sentence summary Tunable organoid modeling and CODA architectural quantification in tandem help design a tissue-validated organoid model.
Collapse
|
16
|
Tezuka M, Tsuchioka K, Kobayashi K, Kuramochi Y, Kiguchi S. Suppressive effects of linzagolix, a novel non-peptide antagonist of gonadotropin-releasing hormone receptors, in experimental endometriosis model rats. Clin Exp Pharmacol Physiol 2023. [PMID: 37186405 DOI: 10.1111/1440-1681.13778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023]
Abstract
Endometriosis is an oestrogen-dependent disease in which endometrial-like tissue grows outside the uterus in women of reproductive age. Accordingly, control of oestradiol (E2) levels is an effective treatment for endometriosis. Because gonadotropin-releasing hormone (GnRH) is the main controller of E2 secretion, control of GnRH signalling by GnRH antagonism is an effective strategy for the treatment of sex hormone-dependent diseases such as endometriosis. The purpose of the present study was to evaluate the effects of the potent, orally available and selective GnRH antagonist linzagolix on experimental endometriosis in rats and compare them with those of dienogest, which is used clinically to treat endometriosis. Experimental endometriosis was induced in female rats at the proestrus stage of the oestrous cycle via autotransplantation of endometrial tissue into the renal subcapsular space. Linzagolix significantly decreased cyst volumes compared with the control group at doses of 50 mg/kg or more. Indeed, a suppressive effect of dienogest on cyst volume was observed only at the highest dose evaluated (1 mg/kg). The effective concentration of linzagolix, calculated as the free form of the last-observed drug concentration, was ~1 μmol/L in endometriosis model rats. The present study also reveals that linzagolix exerts a sustained inhibitory effect on E2 secretion, indicating that the suppressive effect on endometriosis cyst volumes could be attributed to its pharmacological suppression of GnRH signalling and serum E2 concentrations. Altogether, our findings indicate that linzagolix may be a useful therapeutic intervention for hormone-dependent diseases including endometriosis.
Collapse
Affiliation(s)
- Motohiro Tezuka
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Kumi Tsuchioka
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Kaoru Kobayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Yu Kuramochi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Sumiyoshi Kiguchi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| |
Collapse
|
17
|
Ser HL, Au Yong SJ, Shafiee MN, Mokhtar NM, Ali RAR. Current Updates on the Role of Microbiome in Endometriosis: A Narrative Review. Microorganisms 2023; 11:360. [PMID: 36838325 PMCID: PMC9962481 DOI: 10.3390/microorganisms11020360] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/16/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Endometriosis affects approximately 6 to 10% of reproductive-age women globally. Despite much effort invested, the pathogenesis that promotes the development, as well as the progression of this chronic inflammatory disease, is poorly understood. The imbalance in the microbiome or dysbiosis has been implicated in a variety of human diseases, especially the gut microbiome. In the case of endometriosis, emerging evidence suggests that there may be urogenital-gastrointestinal crosstalk that leads to the development of endometriosis. Researchers may now exploit important information from microbiome studies to design endometriosis treatment strategies and disease biomarkers with the use of advanced molecular technologies and increased computational capacity. Future studies into the functional profile of the microbiome would greatly assist in the development of microbiome-based therapies to alleviate endometriosis symptoms and improve the quality of life of women suffering from endometriosis.
Collapse
Affiliation(s)
- Hooi-Leng Ser
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway 47500, Malaysia
| | - Siu-Jung Au Yong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway 47500, Malaysia
| | - Mohamad Nasir Shafiee
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsan Malaysia, Cheras 56000, Malaysia
| | - Norfilza Mohd Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Raja Affendi Raja Ali
- School of Medical and Life Sciences, Sunway University, Bandar Sunway 47500, Malaysia
- Gut Research Group, Faculty of Medicine, Universiti Kebangsan Malaysia, Cheras 56000, Malaysia
| |
Collapse
|
18
|
Rodent Animal Models of Endometriosis-Associated Pain: Unmet Needs and Resources Available for Improving Translational Research in Endometriosis. Int J Mol Sci 2023; 24:ijms24032422. [PMID: 36768741 PMCID: PMC9917069 DOI: 10.3390/ijms24032422] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Chronic pain induced by endometriosis is a maladaptive pain experienced by half of women with this disease. The lack of pharmacological treatments suitable for the long-term relief of endometriosis-associated pain, without an impact on fertility, remains an urgent unmet need. Progress has been slowed by the absence of a reproducible rodent endometriosis model that fully replicates human physiopathological characteristics, including pain symptoms. Although pain assessment in rodents is a complicated task requiring qualified researchers, the choice of the behavioral test is no less important, since selecting inappropriate tests can cause erroneous data. Pain is usually measured with reflex tests in which hypersensitivity is evaluated by applying a noxious stimulus, yet this ignores the associated emotional component that could be evaluated via non-reflex tests. We conducted a systematic review of endometriosis models used in rodents and the number of them that studied pain. The type of behavioral test used was also analyzed and classified according to reflex and non-reflex tests. Finally, we determined the most used reflex tests for the study of endometriosis-induced pain and the main non-reflex behavioral tests utilized in visceral pain that can be extrapolated to the study of endometriosis and complement traditional reflex tests.
Collapse
|
19
|
He Y, Liang B, Hung SW, Zhang R, Xu H, Chung JPW, Wang CC. Re-evaluation of mouse models of endometriosis for pathological and immunological research. Front Immunol 2022; 13:986202. [PMID: 36466829 PMCID: PMC9716019 DOI: 10.3389/fimmu.2022.986202] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/24/2022] [Indexed: 08/30/2023] Open
Abstract
Endometriosis is an estrogen-dependent gynecological disease with chronic pelvic inflammation. In order to study the pathophysiology of endometriosis and examine the therapeutic effects of new pharmaceuticals for endometriosis treatment, different animal models had been developed in the last two decades, especially mouse models. However, no study evaluated the effects of various modeling approaches on pathology and immunology in endometriosis. This study aimed to compare endometriotic lesion development and immune profiles under different methods of establishing endometriosis models in mice, including estrus synchronization (ovariectomy with estrogen supplement versus male urine-soaked transfer bedding), endometrium preparations (whole uterus including endometrium and myometrium fragments versus solely endometrium fragments), and surgical transplantation (subcutaneous transplantation versus intraperitoneal injection). Our results showed that lesion growth under estrus synchronization by ovariectomy with estrogen supplement had a higher success rate and more proliferative endometrium, apart from higher body weight gain. Immune responses in peripheral blood were similar in the whole uterus and solely endometrium fragments and in intraperitoneal injection and subcutaneous transplantation, but a more innate immune response in the peritoneal microenvironment was found in solely endometrium fragments and intraperitoneal injection than counterparts. In conclusion, different endometriosis modeling methods result in different pathological and immunological features. Ovariectomy with estrogen supplement, solely endometrium fragments, and intraperitoneal injection are more suitable for both pathological and immunological studies of endometriosis in mice, which are important for mechanistic studies and immunotherapy development.
Collapse
Affiliation(s)
- Ying He
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Gynecological Oncology, Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Bo Liang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sze Wan Hung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ruizhe Zhang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hui Xu
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jacqueline Pui Wah Chung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Chinese University of Hong Kong, Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
20
|
da Silva FR, Soares Thimoteo D, Ferraz Carbonel A, Fuchs LFP, da Silva Sasso GR, Dos Santos Simões R, Invitti A, Ramos Vieira R, de Souza Ferreira LP, da Silva RA, Lima PDA, Soares Júnior JM, de Jesus Simões M, Bertoncini CRA. Histomorphometric analysis of the endometrium in an ectopic model of endometriosis in mice. Gynecol Endocrinol 2022; 38:874-878. [PMID: 36067792 DOI: 10.1080/09513590.2022.2119218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Objective: Evaluate histomorphometry of ectopic and eutopic endometrial tissues in receptor mice. Method: Eighteen female Balb/c were divided into 3 groups, 6 animals each: GI Control, no procedure; GII - Sham, animals that had the same procedures as GIII without receiving the ectopic endometrial implant. Instead, they received saline solution; GIII - endometriosis model, animals had surgical intervention with an ectopic endometrial implant. GI and GIII mice were treated with 17β-estradiol, 100 µg/kg each. All animals were euthanized to collect uterine horns, which were fixed in 4% paraformaldehyde, embedded in paraffin, stained with Hematoxilin and Eosin and submitted to histomorphometric analyzes. Data underwent one-way ANOVA followed by Tukey's test. Results: Local tissue growth, showing important lesions and adhesions, as well as dark cysts were noticed. In GIII group, there was an increase in number of blood vessels and glands (GIII ≥ GI and GIII p > .001). Thickening of the GIII endometrial epithelial was also evident (GIII ≥ GI and GIII. p > .001). We also noticed an increase in the number of eosinophils (GIII (GIII ≥ GI and GIII. p > .001). Conclusion: Easy to perform model, capable of reproducing morphological endometriosis characteristics. From our findings, there was an increase of endometrial thickness as well as an increase in the eosinophils population.
Collapse
Affiliation(s)
- Felipe Rosendo da Silva
- Department of Gynecology - Paulista School of Medicine, Federal University of São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | - Daniel Soares Thimoteo
- Department of Gynecology - Paulista School of Medicine, Federal University of São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | - Adriana Ferraz Carbonel
- Department of Gynecology - Paulista School of Medicine, Federal University of São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
- Structural and Functional Biology Graduate Program, Paulista School of Medicine, Universidade Federal de São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | - Luiz Fernando Portugal Fuchs
- Department of Gynecology - Paulista School of Medicine, Federal University of São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | - Gisela Rodrigues da Silva Sasso
- Structural and Functional Biology Graduate Program, Paulista School of Medicine, Universidade Federal de São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | - Ricardo Dos Santos Simões
- Department of Obstetrics and Gynecology, Medicine Faculty of University of São Paulo - FMUSP, São Paulo, SP, Brazil
| | - Adriana Invitti
- Department of Gynecology - Paulista School of Medicine, Federal University of São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | - Renata Ramos Vieira
- Department of Gynecology - Paulista School of Medicine, Federal University of São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | - Luiz Philipe de Souza Ferreira
- Structural and Functional Biology Graduate Program, Paulista School of Medicine, Universidade Federal de São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | - Rafael André da Silva
- Biosciences Graduate Program, Institute of Biosciences, Letters and Exact Sciences, São Paulo State University (IBILCE/UNESP), São José do Rio Preto, SP, Brazil
| | | | - José Maria Soares Júnior
- Department of Obstetrics and Gynecology, Medicine Faculty of University of São Paulo - FMUSP, São Paulo, SP, Brazil
| | - Manuel de Jesus Simões
- Department of Gynecology - Paulista School of Medicine, Federal University of São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
- Structural and Functional Biology Graduate Program, Paulista School of Medicine, Universidade Federal de São Paulo - EPM/UNIFESP, São Paulo, SP, Brazil
| | | |
Collapse
|
21
|
Kanellopoulos D, Karagianni D, Pergialiotis V, Nikiteas N, Lazaris AC, Iliopoulos D. The effect of endometriosis on fertility in an animal model. J Med Life 2022; 15:1170-1175. [PMID: 36415526 PMCID: PMC9635238 DOI: 10.25122/jml-2021-0391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/18/2022] [Indexed: 11/21/2022] Open
Abstract
The present experimental model aimed to investigate the possible effect of endometriosis on ovarian function by altering follicular maturation and development. This single-blind, randomized study included twenty-four female Sprague Dawley mice, 2.5 months old, weighing 160-200 grams. The animals were randomly separated into four groups on the day of the surgery. Each group consisted of 6 mice. The first group (A) consisted of healthy female mice (control group). The second group (B) consisted of mice subjected to surgical insertion of ovarian endometrioma. The third group (C) consisted of mice subjected to surgically induced diffuse intraperitoneal endometriosis, and the fourth group (D) consisted of mice subjected to surgically induced extraperitoneal endometriosis. According to our experimental model, endometriosis may affect ovarian function by increasing the number of luteinized unruptured follicles (follicles that have undergone luteinization without prior rupture).
Collapse
Affiliation(s)
- Dimitrios Kanellopoulos
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra Karagianni
- 1 Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasilios Pergialiotis
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Nikiteas
- 2 Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas C Lazaris
- 1 Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Kim TH, Bae N, Kim T, Hsu AL, Hunter MI, Shin JH, Jeong JW. Leptin Stimulates Endometriosis Development in Mouse Models. Biomedicines 2022; 10:biomedicines10092160. [PMID: 36140261 PMCID: PMC9496281 DOI: 10.3390/biomedicines10092160] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/23/2022] [Accepted: 08/27/2022] [Indexed: 11/24/2022] Open
Abstract
Endometriosis is a chronic inflammatory condition in women, and obesity leads to an inflammatory condition that is directly involved in the etiology of endometriosis. However, observational studies have shown an inverse correlation between endometriosis and a low body mass index (BMI). Obesity does not protect against endometriosis, and on the contrary, an increased BMI may lead to more severe forms of the disease. To determine the effect of obesity on endometriosis, diet-induced and genetically engineered obese mouse models were integrated with endometriosis mouse models with fluorescence-tagged ectopic lesions. High-fat diet-induced obese mice revealed a significant increase in endometriosis development compared with regular-diet control mice. However, obese recipient mice with leptin deficiency and leptin receptor deficiency showed suppressed endometriosis development compared with control mice. Furthermore, donor uterine tissues with leptin deficiency and leptin receptor deficiency suppressed endometriosis development compared with control donor in control recipient mice. Importantly, we revealed that aberrant high levels of leptin concentration significantly increased endometriosis development compared with vehicle treatment group in control mice with normal body weight. Our results suggest that leptin and its receptor are critical for endometriosis development.
Collapse
Affiliation(s)
- Tae Hoon Kim
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49534, USA
| | - Nayoung Bae
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Guro Hospital, Korea University Medical Center, Seoul 02841, Korea
| | - Taeho Kim
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Albert L. Hsu
- Reproductive Medicine and Fertility Center, Department of Obstetrics, Gynecology & Women’s Health, University of Missouri School of Medicine, Columbia, MO 65202, USA
| | - Mark I. Hunter
- Department of Obstetrics, Gynecology & Women’s Health, University of Missouri School of Medicine, Columbia, MO 65202, USA
| | - Jung-Ho Shin
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Guro Hospital, Korea University Medical Center, Seoul 02841, Korea
- Correspondence: (J.-H.S.); (J.-W.J.)
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49534, USA
- Correspondence: (J.-H.S.); (J.-W.J.)
| |
Collapse
|
23
|
Maenhoudt N, De Moor A, Vankelecom H. Modeling Endometrium Biology and Disease. J Pers Med 2022; 12:1048. [PMID: 35887546 PMCID: PMC9316888 DOI: 10.3390/jpm12071048] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022] Open
Abstract
The endometrium, lining the uterine lumen, is highly essential for human reproduction. Its exceptional remodeling plasticity, including the transformation process to welcome and nest the embryo, is not well understood. Lack of representative and reliable study models allowing the molecular and cellular mechanisms underlying endometrium development and biology to be deciphered is an important hurdle to progress in the field. Recently, powerful organoid models have been developed that not only recapitulate endometrial biology such as the menstrual cycle, but also faithfully reproduce diseases of the endometrium such as endometriosis. Moreover, single-cell profiling endeavors of the endometrium in health and disease, and of derived organoids, start to provide deeper insight into cellular complexity and expression specificities, and in resulting tissue processes. This granular portrayal will not only help in understanding endometrium biology and disease, but also in pinning down the tissue's stem cells, at present not yet conclusively defined. Here, we provide a general overview of endometrium development and biology, and the efforts of modeling both the healthy tissue, as well as its key diseased form of endometriosis. The future of modeling and deciphering this key tissue, hidden inside the womb, looks bright.
Collapse
Affiliation(s)
| | | | - Hugo Vankelecom
- Unit of Stem Cell Research, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, Leuven Stem Cell Institute, 3000 Leuven, Belgium; (N.M.); (A.D.M.)
| |
Collapse
|
24
|
Tan Z, Hung SW, Zheng X, Wang CC, Chung JPW, Zhang T. What We Have Learned from Animal Models to Understand the Etiology and Pathology of Endometrioma-Related Infertility. Biomedicines 2022; 10:biomedicines10071483. [PMID: 35884788 PMCID: PMC9313443 DOI: 10.3390/biomedicines10071483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Endometrioma (OMA) is the most common subtype of endometriosis, in which the endometriotic lesions are implanted in the ovary. Women with OMA are usually associated with infertility, presenting with reduced ovarian reserve, low oocyte quantity and quality, and poor fertility outcomes. However, the underlying pathological mechanisms in OMA-related infertility are still unclear. Due to the limitations and ethical issues of human studies in reproduction, animal models that recapitulate OMA characteristics and its related infertility are critical for mechanistic studies and subsequent drug development, preclinical testing, and clinical trials. This review summarized the investigations of OMA-related infertility based on previous and latest endometrioma models, providing the possible pathogenesis and potential therapeutic targets for further studies.
Collapse
Affiliation(s)
- Zhouyurong Tan
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong; (Z.T.); (S.-W.H.); (X.Z.); (C.-C.W.); (J.P.-W.C.)
| | - Sze-Wan Hung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong; (Z.T.); (S.-W.H.); (X.Z.); (C.-C.W.); (J.P.-W.C.)
| | - Xu Zheng
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong; (Z.T.); (S.-W.H.); (X.Z.); (C.-C.W.); (J.P.-W.C.)
| | - Chi-Chiu Wang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong; (Z.T.); (S.-W.H.); (X.Z.); (C.-C.W.); (J.P.-W.C.)
- Reproduction and Development, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
- Sichuan University-Chinese University of Hong Kong Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Jacqueline Pui-Wah Chung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong; (Z.T.); (S.-W.H.); (X.Z.); (C.-C.W.); (J.P.-W.C.)
| | - Tao Zhang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong; (Z.T.); (S.-W.H.); (X.Z.); (C.-C.W.); (J.P.-W.C.)
- Correspondence: ; Tel.: +852-3505-3099
| |
Collapse
|
25
|
Kanellopoulos D, Karagianni D, Pergialiotis V, Patsouras G, Patsouras K, Nikiteas N, Lazaris AC, Iliopoulos D. The interplay between endometriosis and fertility in rats: a systematic review. J Med Life 2022; 15:742-746. [PMID: 35928366 PMCID: PMC9321488 DOI: 10.25122/jml-2021-0329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/04/2022] [Indexed: 11/16/2022] Open
Abstract
For the last decades, endometriosis has been a major gynecological problem and a significant cause of infertility for women worldwide. It is estimated that the disease affects about 10-15% of all women of reproductive age and 70% of women suffering from chronic pelvic pain. At the same time, the incidence is about 40-60% in women with dysmenorrhea and 20-30% in women with subfertility. Despite the high percentage of affected women, endometriosis is still characterized by insufficient knowledge of the pathogenic processes, leading to the development and continuity of the disease. For this reason, there is a significant need for insight and understanding of the pathogenesis of endometriosis. This systematic review aims to present the latest data on the use of rats in endometriosis research and to explore how fertility is affected in rats with endometriosis. The methodology included a review of the available publications retrieved by a search in various scientific databases, such as PubMed, Scopus, Medline, and Google Scholar. The initial search generated 30 titles, with 10 articles fulfilling the inclusion criteria. In conclusion, several surgical techniques have been proposed to induce endometriosis, mainly using rats as the appropriate animal model. Studies in rats showed that endometriosis causes infertility and that pregnancy rates are lower for rats with endometriosis than those without endometriosis. In addition, rats with endometriosis have significant abnormalities in the structure of their oocytes as well as in the development of their embryos (genetic abnormalities).
Collapse
Affiliation(s)
- Dimitrios Kanellopoulos
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, National and Kapodistrian University of Athens, Athens, Greece,Department of Obstetrics and Gynecology, Tzaneio Hospital, Athens, Greece,Corresponding Author: Dimitrios Kanellopoulos, Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, National and Kapodistrian University of Athens, Athens, Greece. E-mail:
| | - Dimitra Karagianni
- 1 Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasilios Pergialiotis
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Nikolaos Nikiteas
- 2 Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas C Lazaris
- 1 Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
26
|
Park Y, Demessie AA, Luo A, Taratula OR, Moses AS, Do P, Campos L, Jahangiri Y, Wyatt CR, Albarqi HA, Farsad K, Slayden OD, Taratula O. Targeted Nanoparticles with High Heating Efficiency for the Treatment of Endometriosis with Systemically Delivered Magnetic Hyperthermia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107808. [PMID: 35434932 PMCID: PMC9232988 DOI: 10.1002/smll.202107808] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/01/2022] [Indexed: 05/31/2023]
Abstract
Endometriosis is a devastating disease in which endometrial-like tissue forms lesions outside the uterus. It causes infertility and severe pelvic pain in ≈176 million women worldwide, and there is currently no cure for this disease. Magnetic hyperthermia could potentially eliminate widespread endometriotic lesions but has not previously been considered for treatment because conventional magnetic nanoparticles have relatively low heating efficiency and can only provide ablation temperatures (>46 °C) following direct intralesional injection. This study is the first to describe nanoparticles that enable systemically delivered magnetic hyperthermia for endometriosis treatment. When subjected to an alternating magnetic field (AMF), these hexagonal iron-oxide nanoparticles exhibit extraordinary heating efficiency that is 6.4× greater than their spherical counterparts. Modifying nanoparticles with a peptide targeted to vascular endothelial growth factor receptor 2 (VEGFR-2) enhances their endometriosis specificity. Studies in mice bearing transplants of macaque endometriotic tissue reveal that, following intravenous injection at a low dose (3 mg per kg), these nanoparticles efficiently accumulate in endometriotic lesions, selectively elevate intralesional temperature above 50 °C upon exposure to external AMF, and completely eradicate them with a single treatment. These nanoparticles also demonstrate promising potential as magnetic resonance imaging (MRI) contrast agents for precise detection of endometriotic tissue before AMF application.
Collapse
Affiliation(s)
- Youngrong Park
- College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Ananiya A Demessie
- College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Addie Luo
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, 505 NW 185th Avenue Beaverton, Portland, Oregon, 97006, USA
| | - Olena R Taratula
- College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Abraham S Moses
- College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Peter Do
- College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Leonardo Campos
- Dotter Interventional Institute, Department of Interventional Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon, 97239, USA
| | - Younes Jahangiri
- Dotter Interventional Institute, Department of Interventional Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon, 97239, USA
| | - Cory R Wyatt
- Department of Diagnostic Radiology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, Oregon, 97239, USA
- Advanced Imaging Research Center, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, Oregon, 97239, USA
| | - Hassan A Albarqi
- Department of Pharmaceutics, College of Pharmacy, Najran University, King Abdulaziz Road, Najran, 55461, Saudi Arabia
| | - Khashayar Farsad
- Dotter Interventional Institute, Department of Interventional Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon, 97239, USA
| | - Ov D Slayden
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, 505 NW 185th Avenue Beaverton, Portland, Oregon, 97006, USA
| | - Oleh Taratula
- College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| |
Collapse
|
27
|
Marquardt RM, Nafiujjaman M, Kim TH, Chung SJ, Hadrick K, Kim T, Jeong JW. A Mouse Model of Endometriosis with Nanoparticle Labeling for In Vivo Photoacoustic Imaging. Reprod Sci 2022; 29:2947-2959. [PMID: 35641854 DOI: 10.1007/s43032-022-00980-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Endometriosis is a condition of the female reproductive tract characterized by endometrium-like tissue growing outside the uterus. Though it is a common cause of pelvic pain and infertility, there is currently no reliable noninvasive method to diagnose the presence of endometriosis without surgery, and the pathophysiological mechanisms that lead to the occurrence of symptoms require further inquiry. Due to patient heterogeneity and delayed diagnosis, animal models are commonly used to study the development of endometriosis, but these are costly due to the large number of animals needed to test various treatments and experimental conditions at multiple endpoints. Here, we describe a method for synthesis of multimodal imaging gold-fluorescein isothiocyanate (FITC) nanoparticles with preclinical application via induction of nanoparticle-labeled endometriosis-like lesions in mice. Labeling donor endometrial tissue fragments with gold-FITC nanoparticles prior to induction of endometriosis in recipients enables in vivo detection of the gold-labeled lesions with photoacoustic imaging. The same imaging method can be used to visualize embryos noninvasively in pregnant mice. Furthermore, the conjugated FITC dye on the gold nanoparticles allows easy isolation of labeled lesion tissue under a fluorescence dissection microscope. After dissection, the presence of gold-FITC nanoparticles and endometrium-like histology of lesions can be verified through fluorescence imaging, gold enhancement, and immunostaining. This method for in vivo imaging of endometriosis-like lesions and fluorescence-guided dissection will permit new experimental possibilities for the longitudinal study of endometriosis development and progression as well as endometriosis-related infertility.
Collapse
Affiliation(s)
- Ryan M Marquardt
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA.,Cell and Molecular Biology Program, Michigan State University, College of Natural Science, East Lansing, MI, USA
| | - Md Nafiujjaman
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA
| | - Seock-Jin Chung
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Kay Hadrick
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Taeho Kim
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA.
| |
Collapse
|
28
|
Endometrium as Control of Endometriosis in Experimental Research: Assessment of Sample Suitability. Diagnostics (Basel) 2022; 12:diagnostics12040970. [PMID: 35454018 PMCID: PMC9032605 DOI: 10.3390/diagnostics12040970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Endometriosis is a chronic gynecological disease that causes numerous severe symptoms in affected women. Revealing alterations of the molecular processes in ectopic endometrial tissue is the current policy for understanding the pathomechanisms and discovering potential novel therapeutic targets. Examining molecular processes of eutopic endometrium is likely to be a convenient method to compare it with the molecular alterations observed in ectopic tissues. The aim of the present study was to determine what proportion of the surgically resected eutopic endometrial samples is suitable for further experiments so that these can be comparable with endometriosis. Final hospital reports and histopathology reports of a 3-year-long period (1162 cases) were analysed. The application of a retrospective screening method promoted the categorization of these cases, and quantification of the categorized cases was accomplished. In addition, results obtained from cultured endometrium samples were also detailed. Only a small number of the harvested endometrial samples was suitable for further molecular analysis, while preoperative screening protocol could enlarge this fraction. Applying clinical and histopathological selection and exclusion criteria for tissue screening and histopathological examination of samples could ensure the comparability of healthy endometrium with endometriosis. The present study could be useful for researchers who intend to perform molecular experiments to compare endometriosis with the physiological processes of the endometrium.
Collapse
|
29
|
Yoo JY, Kim TH, Shin JH, Marquardt RM, Müller U, Fazleabas AT, Young SL, Lessey BA, Yoon HG, Jeong JW. Loss of MIG-6 results in endometrial progesterone resistance via ERBB2. Nat Commun 2022; 13:1101. [PMID: 35232969 PMCID: PMC8888616 DOI: 10.1038/s41467-022-28608-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/27/2021] [Indexed: 01/17/2023] Open
Abstract
Female subfertility is highly associated with endometriosis. Endometrial progesterone resistance is suggested as a crucial element in the development of endometrial diseases. We report that MIG-6 is downregulated in the endometrium of infertile women with endometriosis and in a non-human primate model of endometriosis. We find ERBB2 overexpression in the endometrium of uterine-specific Mig-6 knockout mice (Pgrcre/+Mig-6f/f; Mig-6d/d). To investigate the effect of ERBB2 targeting on endometrial progesterone resistance, fertility, and endometriosis, we introduce Erbb2 ablation in Mig-6d/d mice (Mig-6d/dErbb2d/d mice). The additional knockout of Erbb2 rescues all phenotypes seen in Mig-6d/d mice. Transcriptomic analysis shows that genes differentially expressed in Mig-6d/d mice revert to their normal expression in Mig-6d/dErbb2d/d mice. Together, our results demonstrate that ERBB2 overexpression in endometrium with MIG-6 deficiency causes endometrial progesterone resistance and a nonreceptive endometrium in endometriosis-related infertility, and ERBB2 targeting reverses these effects. Female subfertility is highly associated with endometriosis. Here the authors show that progesterone-induced MIG-6 is reduced in endometrium of infertile women and non-human primates with endometriosis, and in a mouse model find that Erbb2 is the key mediator of Mig-6 loss induced endometriosis-related infertility.
Collapse
Affiliation(s)
- Jung-Yoon Yoo
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA.,Department of Biomedical Laboratory Science, Yonsei University Mirae Campus, Wonju, South Korea
| | - Tae Hoon Kim
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA
| | - Jung-Ho Shin
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, Guro Hospital, Korea University Medical Center, Seoul, South Korea
| | - Ryan M Marquardt
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA.,Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Asgerally T Fazleabas
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA
| | - Steven L Young
- Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, NC, USA
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
| | - Jae-Wook Jeong
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA.
| |
Collapse
|
30
|
Matta K, Lefebvre T, Vigneau E, Cariou V, Marchand P, Guitton Y, Royer AL, Ploteau S, Le Bizec B, Antignac JP, Cano-Sancho G. Associations between persistent organic pollutants and endometriosis: A multiblock approach integrating metabolic and cytokine profiling. ENVIRONMENT INTERNATIONAL 2022; 158:106926. [PMID: 34649050 DOI: 10.1016/j.envint.2021.106926] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 10/06/2021] [Indexed: 05/09/2023]
Abstract
Humans are exposed daily to complex mixtures of chemical pollutants through their environment and diet, some of which have the potential to disrupt the bodies' natural endocrine functions and contribute to reproductive diseases like endometriosis. Increasing epidemiological and experimental evidence supports the association between endometriosis and certain persistent organic pollutants (POPs) like dioxins; however, little is known about the underlying linking mechanisms. The main objective of this study is to proof the methodological applicability and discovery potential of integrating ultra-trace mass spectrometry (MS) profiling of POP biomarkers and endogenous biomarker profiling (MS metabolomics and cytokines) in a case-control study for the etiological research of endometriosis. The approach is applied in a pilot clinical-based study conducted in France where women with and without surgically confirmed endometriosis were recruited. Serum samples were analysed with high-resolution MS for about 30 polychlorinated biphenyls (PCBs), organochlorinated pesticides and perfluoroalkyl substances (PFAS). About 600 serum metabolites and lipids were identified with targeted metabolomics using tandem MS with the Biocrates MxP® Quant 500 Kit. A panel of 4 pro-inflammatory cytokines were analysed using ELISA-based 4-PLEX analyser. Statistical analysis included a battery of variable selection approaches, multivariate logistic regression for single-chemical associations, Bayesian kernel machine regressions (BKMR) to identify mixture effects of POPs and a multiblock approach to identify shared biomarker signatures among high risk clusters. The results showed the positive associations between some POPs and endometriosis risk, including the pesticide trans-nonachlor Odds Ratio (95% Confidence Interval) 3.38 (2.06-5.98), p < 0.0001 and PCB 114 OR (95% CI) 1.83 (1.17-2.93), p = 0.009. The BKMR approach showed a tendency of a positive cumulative effect of the mixture, however trans-nonachlor exhibited significant associations within the mixture and interacted with other PCBs, strengthening the effects at highest concentrations. Finally, the multiblock analysis, relating the various blocks of data, revealed a latent cluster of women with higher risk of endometrioma presenting higher concentrations of trans-nonachlor, PCB 114 and dioxin-like toxic equivalents from PCBs, together with an increased inflammatory profile (i.e. elevated interleukin-8 and monocyte chemoattractant protein-1). It was also highlighted a specific metabolic pattern characterized by dysregulation of bile acid homeostasis and lipase activity. Further research will be required with larger sample size to confirm these findings and gain insight on the underlying mechanisms between POPs and endometriosis.
Collapse
Affiliation(s)
| | - Tiphaine Lefebvre
- LABERCA, Oniris, INRAE, 44307 Nantes, France; Faculty of Medicine, University of Nantes, Nantes, France; Department of Biology and Reproductive Medicine, University Hospital of Nantes, Nantes, France
| | | | | | | | | | | | - Stéphane Ploteau
- Faculty of Medicine, University of Nantes, Nantes, France; Service de Gynecologie-obstétrique, CIC FEA, Hôpital Mère Enfant, CHU Hôtel Dieu, Nantes, France
| | | | | | | |
Collapse
|
31
|
Lee JH, Park JH, Won BH, Im W, Cho S. Administration of red ginseng regulates microRNA expression in a mouse model of endometriosis. Clin Exp Reprod Med 2021; 48:337-346. [PMID: 34781599 PMCID: PMC8651755 DOI: 10.5653/cerm.2021.04392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/20/2021] [Accepted: 07/12/2021] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Red ginseng (RG) exerts anti-inflammatory, anti-proliferative, and immunomodulatory effects on endometriosis through the regulation of microRNA (miRNA) expression. It may also ameliorate endometriosis by affecting the expression of multiple miRNAs simultaneously, rather than acting on a single miRNA at a given time. Since studies on the overall effects of RG on endometriosis via the regulation of miRNA expression are lacking, the current study aimed to explore the global effect of RG on miRNA expression in a mouse model of endometriosis. METHODS To establish the mouse model, the uterine horn of donor mice was implanted into the lateral side of the recipients' peritoneum, followed by vehicle or RG treatment for 8 weeks. RESULTS To confirm the effects of RG on the established mouse model, the size of the implanted uterus was measured; it was found to be lower in mice from the RG group than in mice from the control group. miRNA expression profiles in the implanted uterus of the mouse model of endometriosis after vehicle or RG administration were analyzed using microarray technology. Thereafter, seven candidate miRNAs and 125 candidate genes (miRNA targets) were identified through a bioinformatics analysis. CONCLUSION The present findings suggest that RG regulates the expression of multiple miRNAs and mRNAs, thereby alleviating endometriosis in a mouse model of the disease.
Collapse
Affiliation(s)
- Jae Hoon Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyun Park
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Bo Hee Won
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Wooseok Im
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - SiHyun Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
32
|
Stejskalová A, Vankelecom H, Sourouni M, Ho MY, Götte M, Almquist BD. In vitro modelling of the physiological and diseased female reproductive system. Acta Biomater 2021; 132:288-312. [PMID: 33915315 DOI: 10.1016/j.actbio.2021.04.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
The maladies affecting the female reproductive tract (FRT) range from infections to endometriosis to carcinomas. In vitro models of the FRT play an increasingly important role in both basic and translational research, since the anatomy and physiology of the FRT of humans and other primates differ significantly from most of the commonly used animal models, including rodents. Using organoid culture to study the FRT has overcome the longstanding hurdle of maintaining epithelial phenotype in culture. Both ECM-derived and engineered materials have proved critical for maintaining a physiological phenotype of FRT cells in vitro by providing the requisite 3D environment, ligands, and architecture. Advanced materials have also enabled the systematic study of factors contributing to the invasive metastatic processes. Meanwhile, microphysiological devices make it possible to incorporate physical signals such as flow and cyclic exposure to hormones. Going forward, advanced materials compatible with hormones and optimised to support FRT-derived cells' long-term growth, will play a key role in addressing the diverse array of FRT pathologies and lead to impactful new treatments that support the improvement of women's health. STATEMENT OF SIGNIFICANCE: The female reproductive system is a crucial component of the female anatomy. In addition to enabling reproduction, it has wide ranging influence on tissues throughout the body via endocrine signalling. This intrinsic role in regulating normal female biology makes it susceptible to a variety of female-specific diseases. However, the complexity and human-specific features of the reproductive system make it challenging to study. This has spurred the development of human-relevant in vitro models for helping to decipher the complex issues that can affect the reproductive system, including endometriosis, infection, and cancer. In this Review, we cover the current state of in vitro models for studying the female reproductive system, and the key role biomaterials play in enabling their development.
Collapse
|
33
|
Balasubramanian V, Saravanan R, Joseph LD, Dev B, Gouthaman S, Srinivasan B, Dharmarajan A, Rayala SK, Venkatraman G. Molecular dysregulations underlying the pathogenesis of endometriosis. Cell Signal 2021; 88:110139. [PMID: 34464692 DOI: 10.1016/j.cellsig.2021.110139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/07/2023]
Abstract
Endometriosis is a crippling disease characterized by the presence of endometrium-like tissue or scar outside the uterine cavity, commonly confined to the peritoneal and serosal surfaces of the pelvic organs. 10-15% of women in reproductive age are estimated to be affected by endometriosis. Most of these patients present with infertility and suffer from pelvic pain. The benign disease rarely progresses to malignancy. Regardless of its high prevalence, the pathogenesis of the disease is not fully understood. Treatment options for endometriosis are limited and are often based on a symptomatic approach. The unavailability of proper diagnostic approaches, fewer therapeutic options, and sparse understanding of molecular alterations are responsible for the continued disease burden. Exploring the molecular elements causing the pathogenesis of endometriosis may lead to a number of breakthroughs in the treatment of the illness, such as the discovery of new biomarkers for diagnosis and therapeutic targets that can be a guide to better prognosis and reduced recurrence. The goal of this review is to provide the reader a critical understanding of the disease by summarizing the genetic, immunological, hormonal, and epigenetic deregulations that support the molecular basis for development of endometriotic cyst, with a special focus on the study models needed to analyze these changes in the endometriotic microenvironment.
Collapse
Affiliation(s)
- Vaishnavi Balasubramanian
- Department of Human Genetics, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Roshni Saravanan
- Department of Human Genetics, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Leena Dennis Joseph
- Department of Pathology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Bhawna Dev
- Department of Radiology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Shanmugasundaram Gouthaman
- Department of Surgical Oncology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Bhuvana Srinivasan
- Department of Obstetrics and Gynecology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Human Genetics, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India.
| |
Collapse
|
34
|
Nenicu A, Yordanova K, Gu Y, Menger MD, Laschke MW. Differences in growth and vascularization of ectopic menstrual and non-menstrual endometrial tissue in mouse models of endometriosis. Hum Reprod 2021; 36:2202-2214. [PMID: 34109385 DOI: 10.1093/humrep/deab139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/29/2021] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Is there a difference in the growth and vascularization between murine endometriotic lesions originating from menstrual or non-menstrual endometrial fragments? SUMMARY ANSWER Endometriotic lesions developing from menstrual and non-menstrual tissue fragments share many similarities, but also exhibit distinct differences in growth and vascularization, particularly under exogenous estrogen stimulation. WHAT IS KNOWN ALREADY Mouse models are increasingly used in endometriosis research. For this purpose, menstrual or non-menstrual endometrial fragments serve for the induction of endometriotic lesions. So far, these two fragment types have never been directly compared under identical experimental conditions. STUDY DESIGN, SIZE, DURATION This was a prospective experimental study in a murine peritoneal and dorsal skinfold chamber model of endometriosis. Endometrial tissue fragments from menstruated (n = 15) and non-menstruated (n = 21) C57BL/6 mice were simultaneously transplanted into the peritoneal cavity or dorsal skinfold chamber of non-ovariectomized (non-ovx, n = 17), ovariectomized (ovx, n = 17) and ovariectomized, estrogen-substituted (ovx+E2, n = 17) recipient animals and analyzed throughout an observation period of 28 and 14 days, respectively. PARTICIPANTS/MATERIALS, SETTING, METHODS The engraftment, growth and vascularization of the newly developing endometriotic lesions were analyzed by means of high-resolution ultrasound imaging, intravital fluorescence microscopy, histology and immunohistochemistry. MAIN RESULTS AND THE ROLE OF CHANCE Menstrual and non-menstrual tissue fragments developed into peritoneal endometriotic lesions without differences in growth, microvessel density and cell proliferation in non-ovx mice. Lesion formation out of both fragment types was markedly suppressed in ovx mice. In case of non-menstrual tissue fragments, this effect could be reversed by estrogen supplementation. In contrast, endometriotic lesions originating from menstrual tissue fragments exhibited a significantly smaller volume in ovx+E2 mice, which may be due to a reduced hormone sensitivity. Moreover, menstrual tissue fragments showed a delayed vascularization and a reduced blood perfusion after transplantation into dorsal skinfold chambers when compared to non-menstrual tissue fragments, indicating different vascularization modes of the two fragment types. To limit the role of chance, the experiments were conducted under standardized laboratory conditions. Statistical significance was accepted for a value of P < 0.05. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Endometriotic lesions were induced by syngeneic tissue transplantation into recipient mice without the use of pathological endometriotic tissue of human nature. Therefore, the results obtained in this study may not fully relate to human patients with endometriosis. WIDER IMPLICATIONS OF THE FINDINGS The present study significantly contributes to the characterization of common murine endometriosis models. These models represent important tools for studies focusing on the basic mechanisms of endometriosis and the development of novel therapeutic strategies for the treatment of this frequent gynecological disease. The presented findings indicate that the combination of different experimental models and approaches may be the most appropriate strategy to study the pathophysiology and drug sensitivity of a complex disease such as endometriosis under preclinical conditions. STUDY FUNDING/COMPETING INTEREST(S) There was no specific funding of this study. The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- A Nenicu
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - K Yordanova
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Y Gu
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - M D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - M W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
35
|
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
|
36
|
Matta K, Koual M, Ploteau S, Coumoul X, Audouze K, Le Bizec B, Antignac JP, Cano-Sancho G. Associations between Exposure to Organochlorine Chemicals and Endometriosis: A Systematic Review of Experimental Studies and Integration of Epidemiological Evidence. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:76003. [PMID: 34310196 PMCID: PMC8312885 DOI: 10.1289/ehp8421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 05/04/2021] [Accepted: 06/21/2021] [Indexed: 05/19/2023]
Abstract
BACKGROUND Growing epidemiological evidence suggests that organochlorine chemicals (OCCs), including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), may play a role in the pathogenesis of endometriosis. OBJECTIVES We aimed to systematically review the experimental evidence (in vivo and in vitro) on the associations between exposure to OCCs and endometriosis-related end points. METHODS A systematic review protocol was developed following the National Toxicology Program /Office of Health Assessment and Translation (NTP/OHAT) framework and managed within a web-based interface. In vivo studies designed to evaluate the impact of OCCs on the onset or progression of endometriosis and proliferation of induced endometriotic lesions were eligible. Eligible in vitro studies included single-cell and co-culture models to evaluate the proliferation, migration, and/or invasion of endometrial cells. We applied the search strings to PubMed, Web of Science, and Scopus®. A final search was performed on 24 June 2020. Assessment of risk of bias and the level of evidence and integration of preevaluated epidemiological evidence was conducted using NTP/OHAT framework Results: Out of 812 total studies, 39 met the predetermined eligibility criteria (15 in vivo, 23 in vitro, and 1 both). Most studies (n=27) tested TCDD and other dioxin-like chemicals. In vivo evidence supported TCDD's promotion of endometriosis onset and lesion growth. In vitro evidence supported TCDD's promotion of cell migration and invasion, but there was insufficient evidence for cell proliferation. In vitro evidence further supported the roles of the aryl hydrocarbon receptor and matrix metalloproteinases in mediating steroidogenic disruption and inflammatory responses. Estrogen interactions were found across studies and end points. CONCLUSION Based on the integration of a high level of animal evidence with a moderate level of epidemiological evidence, we concluded that TCDD was a known hazard for endometriosis in humans and the conclusion is supported by mechanistic in vitro evidence. Nonetheless, there is need for further research to fill in our gaps in understanding of the relationship between OCCs and their mixtures and endometriosis, beyond the prototypical TCDD. https://doi.org/10.1289/EHP8421.
Collapse
Affiliation(s)
- Komodo Matta
- Oniris, INRAE, UMR 1329 Laboratoire d’Étude des Résidus et Contaminants dans les Aliments (LABERCA), Nantes, France
| | - Meriem Koual
- Université de Paris, T3S, Institut national de la santé et de la recherche médicale (Inserm) UMR S-1124, Paris, France
- Service de Chirurgie Cancérologique Gynécologique et du Sein, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, Paris, France
| | - Stéphane Ploteau
- Service de gynécologie-obstétrique, Centre d’investigation clinique–Femme Enfant Adolescent, Hôpital Mère Enfant, Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - Xavier Coumoul
- Université de Paris, T3S, Institut national de la santé et de la recherche médicale (Inserm) UMR S-1124, Paris, France
| | - Karine Audouze
- Université de Paris, T3S, Institut national de la santé et de la recherche médicale (Inserm) UMR S-1124, Paris, France
| | - Bruno Le Bizec
- Oniris, INRAE, UMR 1329 Laboratoire d’Étude des Résidus et Contaminants dans les Aliments (LABERCA), Nantes, France
| | - Jean-Philippe Antignac
- Oniris, INRAE, UMR 1329 Laboratoire d’Étude des Résidus et Contaminants dans les Aliments (LABERCA), Nantes, France
| | - German Cano-Sancho
- Oniris, INRAE, UMR 1329 Laboratoire d’Étude des Résidus et Contaminants dans les Aliments (LABERCA), Nantes, France
| |
Collapse
|
37
|
Organ-on-a-chip technology for the study of the female reproductive system. Adv Drug Deliv Rev 2021; 173:461-478. [PMID: 33831478 DOI: 10.1016/j.addr.2021.03.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/11/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
Over the past decade, organs-on-a-chip and microphysiological systems have emerged as a disruptive in vitro technology for biopharmaceutical applications. By enabling new capabilities to engineer physiological living tissues and organ units in the precisely controlled environment of microfabricated devices, these systems offer great promise to advance the frontiers of basic and translational research in biomedical sciences. Here, we review an emerging body of interdisciplinary work directed towards harnessing the power of organ-on-a-chip technology for reproductive biology and medicine. The focus of this topical review is to provide an overview of recent progress in the development of microengineered female reproductive organ models with relevance to drug delivery and discovery. We introduce the engineering design of these advanced in vitro systems and examine their applications in the study of pregnancy, infertility, and reproductive diseases. We also present two case studies that use organ-on-a-chip design principles to model placental drug transport and hormonally regulated crosstalk between multiple female reproductive organs. Finally, we discuss challenges and opportunities for the advancement of reproductive organ-on-a-chip technology.
Collapse
|
38
|
Ortiz CN, Torres-Reverón A, Appleyard CB. Metabolomics in endometriosis: challenges and perspectives for future studies. REPRODUCTION AND FERTILITY 2021; 2:R35-R50. [PMID: 35128453 PMCID: PMC8812441 DOI: 10.1530/raf-20-0047] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Endometriosis is a complex disease characterized by inflammation and the growth of endometrial- like glands and stroma outside the uterine cavity. The pathophysiology of endometriosis is not entirely understood, however, with a prevalence of ~10% of women in their reproductive years, the disease symptoms significantly affect the quality of life of millions of women globally. Metabolomic studies have previously identified specific metabolites that could be a signature of endometriosis. This approach could potentially be used as a non-invasive tool for early diagnosis and provide a better understanding of endometriosis pathophysiology. This review aims to provide insight as to how endometriosis affects the metabolome by reviewing different studies that have used this approach to design follow-up studies. The search query included the term 'endometriosis' in combination with 'metabolomics', 'lipidomics', or 'sphingolipidomics' published between 2012 and 2020. We included studies in humans and animal models. Most studies reported differences in the metabolome of subjects with endometriosis in comparison to healthy controls and used samples taken from serum, endometrial tissue, follicular fluid, urine, peritoneal fluid, or endometrial fluid. Statistically significant metabolites contributed to group separation between patients and healthy controls. Reported metabolites included amino acids, lipids, organic acids, and other organic compounds. Differences in methods, analytical techniques, and the presence of confounding factors can interfere with results and interpretation of data. Metabolomics seems to be a promising tool for identifying significant metabolites in patients with endometriosis. Nonetheless, more investigation is needed in order to understand the significance of the study results.
Lay summary
Endometriosis is a chronic disease affecting the quality of life in one out of every ten women during their reproductive years, causing pain and infertility. It is characterized by inflammation and growth of tissue like the endometrium (uterus lining) outside the uterine cavity. Studies have searched for a predictor of endometriosis-associated changes by observing small molecules necessary for metabolism on a large scale (metabolomics). Metabolomics could serve to resolve one of the biggest challenges that patients with endometriosis face: a delay in diagnosis. In this review, the authors summarize identified potential biomarkers from various bodily fluids and tissues that are characteristic of metabolic processes observed in endometriosis. Biomarkers include cell growth, cell survival, high energy demand, oxidative stress, and fatty acid levels. A metabolomics approach offers promise as a non-invasive tool to identify significant metabolite changes in patients with endometriosis, potentially leading to earlier diagnoses and new opportunities for back-translational strategies.
Collapse
Affiliation(s)
- Camila N Ortiz
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, USA
| | | | - Caroline B Appleyard
- Department of Basic Sciences, Ponce Health Sciences University, Ponce Research Institute, Ponce, Puerto Rico, USA
- Department of Internal Medicine, Ponce Health Sciences University, Ponce, Puerto Rico, USA
| |
Collapse
|
39
|
Simopoulou M, Rapani A, Grigoriadis S, Pantou A, Tsioulou P, Maziotis E, Tzanakaki D, Triantafyllidou O, Kalampokas T, Siristatidis C, Bakas P, Vlahos N. Getting to Know Endometriosis-Related Infertility Better: A Review on How Endometriosis Affects Oocyte Quality and Embryo Development. Biomedicines 2021; 9:273. [PMID: 33803376 PMCID: PMC7998986 DOI: 10.3390/biomedicines9030273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/28/2021] [Accepted: 03/05/2021] [Indexed: 12/20/2022] Open
Abstract
Endometriosis-related infertility describes a case of deteriorated fecundity when endometriosis is diagnosed. Numerous mechanisms have been proposed in an effort to delineate the multifaceted pathophysiology that induces impairment of reproductive dynamics in patients with endometriosis. In this critical analysis, authors present the plethora of molecular events that are entailed and elaborate on how they potentially impair the oocyte's and embryo's competence in patients with endometriosis. Reactive oxygen species, dysregulation of the immune system and cellular architectural disruption constitute the crucial mechanisms that detrimentally affect oocyte and embryo developmental potential. The molecular level impairment of the reproductive tissue is discussed, since differentiation, proliferation and apoptosis constitute focal regulatory cellular functions that appear severely compromised in cases of endometriosis. Mapping the precise molecular mechanisms entailed in endometriosis-related infertility may help delineate the complex nature of the disorder and bring us a step closer to a more personalized approach in understanding, diagnosing and managing endometriosis-related infertility.
Collapse
Affiliation(s)
- Mara Simopoulou
- Laboratory of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias, 11527 Athens, Greece; (A.R.); (S.G.); (A.P.); (P.T.); (E.M.)
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Anna Rapani
- Laboratory of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias, 11527 Athens, Greece; (A.R.); (S.G.); (A.P.); (P.T.); (E.M.)
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Sokratis Grigoriadis
- Laboratory of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias, 11527 Athens, Greece; (A.R.); (S.G.); (A.P.); (P.T.); (E.M.)
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Agni Pantou
- Laboratory of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias, 11527 Athens, Greece; (A.R.); (S.G.); (A.P.); (P.T.); (E.M.)
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli, 15232 Athens, Greece
| | - Petroula Tsioulou
- Laboratory of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias, 11527 Athens, Greece; (A.R.); (S.G.); (A.P.); (P.T.); (E.M.)
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Evangelos Maziotis
- Laboratory of Physiology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias, 11527 Athens, Greece; (A.R.); (S.G.); (A.P.); (P.T.); (E.M.)
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Despina Tzanakaki
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Olga Triantafyllidou
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Theodoros Kalampokas
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Charalampos Siristatidis
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Panagiotis Bakas
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| | - Nikolaos Vlahos
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 76, Vasilisis Sofias Avenue, 11528 Athens, Greece; (D.T.); (O.T.); (T.K.); (C.S.); (P.B.); (N.V.)
| |
Collapse
|
40
|
Bora G, Yaba A. The role of mitogen-activated protein kinase signaling pathway in endometriosis. J Obstet Gynaecol Res 2021; 47:1610-1623. [PMID: 33590617 DOI: 10.1111/jog.14710] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/10/2021] [Accepted: 01/29/2021] [Indexed: 12/20/2022]
Abstract
AIM Endometriosis is an estrogen-dependent chronic inflammatory condition which causes pain, infertility, and predisposition for ovarian cancer. Endometriosis generates a unique microenvironment for survivability of endometriotic lesions which includes cell proliferation, differentiation, migration, and apoptosis. For these cellular activities, cascading activations of intracellular kinases are needed. Many kinase signaling pathways, IKKβ/NK-κB pathway, PI3K/AKT/mTOR, and the mitogen-activated protein kinase (MAPK) pathways (ERK1/2, p38, and JNK), are activated in endometriosis. In this review, we focus on the role of MAPK pathways in endometriosis. METHODS To identify the role of MAP Kinase signaling pathway in endometriosis we searched the Pubmed database using the search terms in various combinations "endometriosis," "endometrium," "ovary," "MAPK pathway," "ERK pathway," "p38 pathway," "JNK pathway," "estrogen," and "progesterone." RESULTS According to the current literature, MAPK signaling pathway has various roles in generating microenvironment and survival of endometriosis. Abnormal MAPK activation in migration, implantation, growth, invasion into the pelvic structures, proliferation, and apoptosis leads to the form of endometriosis and to worsen the condition in patients with endometriosis. CONCLUSION To further investigations on the effective and long-term endometriosis treatment, MAPK signaling pathways may be targeted. Molecular mechanism of MAPK signaling pathway in endometriosis should be more deeply understood and clinical trials should be more commonly performed for possible new endometriosis treatments to improve fertility and rescue endometriosis irreversibly.
Collapse
Affiliation(s)
- Gizem Bora
- Department of Histology and Embryology, Yeditepe University School of Medicine, İstanbul, Turkey
| | - Aylin Yaba
- Department of Histology and Embryology, Yeditepe University School of Medicine, İstanbul, Turkey
| |
Collapse
|
41
|
Wuyung PE, Rahadiati FB, Tjahjadi H, Salinah S, Kusmardi K, Kodariah R, Wiweko B. Histopathology and ARID1A Expression in Endometriosis- Associated Ovarian Carcinoma (EAOC) Carcinogenesis Model with Endometrial Autoimplantation and DMBA Induction. Asian Pac J Cancer Prev 2021; 22:553-558. [PMID: 33639673 PMCID: PMC8190335 DOI: 10.31557/apjcp.2021.22.2.553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Ovarian carcinoma is one of the most deadly malignancies in the gynecologic field. The cause is not yet known, and the clinical symptoms are not specific. Endometrioid carcinoma and ovarian clear cell carcinoma can originate from endometriosis and are known as endometriosis-related ovarian carcinoma (EAOC). Development of EAOC experimental animal models is needed for basic research and clinical preparation of human tissue tests. This study aimed to determine the role of the ARID1A gene mutation in the carcinogenetic process of EAOC in experimental animal models induced with DMBA. METHODS In this study, the EAOC experimental model was developed using the autoimplantation technique and DMBA induction. This study involved placebo surgery mice (sham), endometrial autoimplantation, and a combination of endometrial autoimplantation and DMBA induction, which were sacrificed at weeks 5, 10, and 20, respectively. Histopathological assessment and immunohistochemical ARID1A staining with an assessment of positive percentages were carried out on 200 cells. RESULTS This study produced 1 (20%) atypical endometriosis and 1 (20%) clear cell carcinoma at implantation and after 10 weeks of DMBA induction, and 100% endometrioid carcinoma in the DMBA-induced group. ARID1A staining did not show any significant difference (p = 0.313) in all groups. CONCLUSION The combination of endometrial autoimplantation techniques and DMBA induction in the ovary produced atypical endometriosis, clear cell carcinoma, and endometrioid carcinoma, where time is an important factor. There was no significant difference in ARID1A expression between the treatment and control groups.
Collapse
Affiliation(s)
- Puspita Eka Wuyung
- Department of Anatomical Pathology, Faculty of Medicine Universitas Indonesia.,Animal Research Facilities, Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia
| | - Familia Bella Rahadiati
- Specialty Programme in Anatomical Pathology, Department of Anatomical Pathology, Faculty of Medicine Universitas Indonesia
| | - Hartono Tjahjadi
- Department of Anatomical Pathology, Faculty of Medicine Universitas Indonesia
| | - Salinah Salinah
- Department of Anatomical Pathology, Faculty of Medicine Universitas Indonesia
| | - Kusmardi Kusmardi
- Department of Anatomical Pathology, Faculty of Medicine Universitas Indonesia
| | - Ria Kodariah
- Department of Anatomical Pathology, Faculty of Medicine Universitas Indonesia
| | - Budi Wiweko
- Department of Obstetrics and Gynecology, Faculty of Medicine Universitas Indonesia.,Human Reproduction, Infertility, and Family Planning, Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia
| |
Collapse
|
42
|
Tu J, Yang H, Chen Y, Chen Y, Chen H, Li Z, Li L, Zhang Y, Chen X, Yu Z. Current and Future Roles of Circular RNAs in Normal and Pathological Endometrium. Front Endocrinol (Lausanne) 2021; 12:668073. [PMID: 34122342 PMCID: PMC8187767 DOI: 10.3389/fendo.2021.668073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/11/2021] [Indexed: 01/20/2023] Open
Abstract
The uterine endometrium, which lines the mammalian uterus, is essential for embryo implantation. This lining undergoes significant changes during sexual and menstrual cycles. The endometrium is also associated with hormone-related diseases such as endometriosis and endometrial cancer. Circular RNAs (circRNAs) play a role in various biological processes. Recent studies have determined that circRNAs function in both normal and pathological endometrial environments. Here, we review high-throughput studies pertaining to circRNAs as well as individual circRNAs active in the endometrium, in order to explore the myriad functions of circRNAs in the endometrium and mechanisms underlying these functions, from panoramic and individual perspectives. Owing to their abundant expression, stability, and small size, circRNAs have displayed potential usefulness as diagnostic markers and treatment targets for endometrial-related diseases. Therefore, the specific role of circRNAs in the endometrium warrants systematic investigation in the future.
Collapse
Affiliation(s)
- Jiajie Tu
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
- *Correspondence: Jiajie Tu, ; Zhiying Yu,
| | - Huan Yang
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yu Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu Chen
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - He Chen
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhe Li
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Lei Li
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yuanyuan Zhang
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaochun Chen
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhiying Yu
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- *Correspondence: Jiajie Tu, ; Zhiying Yu,
| |
Collapse
|
43
|
Escudero-Lara A, Cabañero D, Maldonado R. Surgical Induction of Endometriosis in Female Mice. Bio Protoc 2020; 10:e3763. [PMID: 33659421 DOI: 10.21769/bioprotoc.3763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/24/2020] [Accepted: 08/17/2020] [Indexed: 11/02/2022] Open
Abstract
Endometriosis is a common gynecological disease characterized by the presence of endometrial tissue outside the uterine cavity. It is frequently associated with pain, infertility and a reduced quality of life, and it lacks adequate treatment. Several rodent models of endometriosis have been developed through heterologous and homologous transplantation of endometrial tissue into the abdominal compartment. Here we describe a surgical procedure to generate a syngeneic model of endometriosis in immunocompetent mice with intact uterine and ovarian tissues. In this model, four uterine fragments from a donor mouse at diestrus are sutured to the abdominal wall of a recipient mouse. One month after surgeries, endometrial implants develop into cysts with glandular epithelium and stroma, mimicking the endometriotic lesions observed in women with endometriosis. Therefore, this mouse model provides a valuable tool to study the pathophysiology of endometriosis and the efficacy of potential treatments.
Collapse
Affiliation(s)
- Alejandra Escudero-Lara
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Universidad Miguel Hernández. Elche, Alicante, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
44
|
Malvezzi H, Marengo EB, Podgaec S, Piccinato CDA. Endometriosis: current challenges in modeling a multifactorial disease of unknown etiology. J Transl Med 2020; 18:311. [PMID: 32787880 PMCID: PMC7425005 DOI: 10.1186/s12967-020-02471-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Endometriosis is a chronic inflammatory hormone-dependent condition associated with pelvic pain and infertility, characterized by the growth of ectopic endometrium outside the uterus. Given its still unknown etiology, treatments usually aim at diminishing pain and/or achieving pregnancy. Despite some progress in defining mode-of-action for drug development, the lack of reliable animal models indicates that novel approaches are required. The difficulties inherent to modeling endometriosis are related to its multifactorial nature, a condition that hinders the recreation of its pathology and the identification of clinically relevant metrics to assess drug efficacy. In this review, we report and comment endometriosis models and how they have led to new therapies. We envision a roadmap for endometriosis research, integrating Artificial Intelligence, three-dimensional cultures and organ-on-chip models as ways to achieve better understanding of physiopathological features and better tailored effective treatments.
Collapse
Affiliation(s)
- Helena Malvezzi
- Hospital Israelita Albert Einstein, São Paulo, SP 05652-900 Brazil
| | - Eliana Blini Marengo
- Instituto Butanta- EstabilidadeBiotech Quality Control, São Paulo, SP 05503-900 Brazil
| | - Sérgio Podgaec
- Hospital Israelita Albert Einstein, São Paulo, SP 05652-900 Brazil
| | | |
Collapse
|
45
|
Gu ZY, Jia SZ, Liu S, Leng JH. Endometrial Organoids: A New Model for the Research of Endometrial-Related Diseases†. Biol Reprod 2020; 103:918-926. [PMID: 32697306 PMCID: PMC7609820 DOI: 10.1093/biolre/ioaa124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/25/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
An ideal research model plays a vital role in studying the pathogenesis of a disease. At present, the most widely used endometrial disease models are cell lines and animal models. As a novel studying model, organoids have already been applied for the study of various diseases, such as disorders related to the liver, small intestine, colon, and pancreas, and have been extended to the endometrium. After a long period of exploration by predecessors, endometrial organoids (EOs) technology has gradually matured and maintained genetic and phenotypic stability after long-term expansion. Compared with cell lines and animal models, EOs have high stability and patient specificity. These not only effectively and veritably reflects the pathophysiology of a disease, but also can be used in preclinical drug screening, combined with patient derived xenografts (PDXs). Indeed, there are still many limitations for EOs. For example, the co-culture system of EOs with stromal cells, immune cell, or vascular cells is not mature, and endometrial cancer organoids have a lower success rate, which should be improved in the future. The investigators predict that EOs will play a significant role in the study of endometrium-related diseases.
Collapse
Affiliation(s)
- Zhi-Yue Gu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Shuang-Zheng Jia
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Song Liu
- Department of Central Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jin-Hua Leng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
46
|
Appleyard CB, Flores I, Torres-Reverón A. The Link Between Stress and Endometriosis: from Animal Models to the Clinical Scenario. Reprod Sci 2020; 27:1675-1686. [PMID: 32542543 DOI: 10.1007/s43032-020-00205-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022]
Abstract
There is strong evidence from humans and animal models showing that abnormal functioning of the hypothalamic-pituitary-adrenal (HPA) axis and/or the inflammatory response system disrupts feedback regulation of both neuroendocrine and immune systems, contributing to disease. Stress is known to affect the physiology of pelvic organs and to disturb the HPA axis leading to chronic, painful, inflammatory disorders. A link between stress and disease has already been documented for many chronic conditions. Endometriosis is a complex chronic gynecological disease associated with severe pelvic pain and infertility that affects 10% of reproductive-aged women. Patients report the negative impact of endometriosis symptoms on quality of life, work/study productivity, and personal relationships, which in turn cause high levels of psychological and emotional distress. The relationship between stress and endometriosis is not clear. Still, we have recently demonstrated that stress increases the size and severity of the lesions as well as inflammatory parameters in an animal model. Furthermore, the "controllability" of stress influences the pathophysiology in this model, offering the possibility of using stress management techniques in patients. The crosstalk between stress-inflammation-pain through HPA axis activity indicates that stress relief should alleviate inflammation and, in turn, decrease painful responses. This opens up the opportunity of altering brain-body-brain pathways as potential new therapeutic option for endometriosis. The goal of this review is to gather the research evidence regarding the interaction between stress (psychological and physiological) and the development and progression of endometriosis on the exacerbation of its symptoms with the purpose of proposing new lines of emerging research and possible treatment modalities for this still incurable disease.
Collapse
Affiliation(s)
- Caroline B Appleyard
- Department of Basic Sciences, Women's Health Division, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA. .,Department of Internal Medicine, Ponce Health Sciences University, Ponce, PR, USA. .,Department of Basic Sciences, Physiology Division, Medical School and Ponce Research Institute, Ponce Health Sciences University, 395 Zona Ind Reparada 2, Ponce, PR, 00716-2347, USA.
| | - Idhaliz Flores
- Department of Basic Sciences, Women's Health Division, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA.,Department of Obstetrics and Gynecology, Ponce Health Sciences University, Ponce, PR, USA
| | | |
Collapse
|
47
|
Zheng P, Jia S, Guo D, Chen S, Zhang W, Cheng A, Xie W, Sun G, Leng J, Lang J. Central Sensitization-Related Changes in Brain Function Activity in a Rat Endometriosis-Associated Pain Model. J Pain Res 2020; 13:95-107. [PMID: 32021399 PMCID: PMC6968808 DOI: 10.2147/jpr.s232313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/17/2019] [Indexed: 12/31/2022] Open
Abstract
Background Pain sensitization processing in the central nervous system may be related to endometriosis-associated pain in patients. The purpose of this study was to understand the alterations in the abnormal pain response in central brain areas and explore the central sensitization mechanism of endometriosis-associated pain. Methods An endometriosis model was established in 40 Sprague-Dawley rats, and the rats underwent pain model assessment through behavioral tests. Twenty Sprague-Dawley rats underwent a sham operation as the control group. Thirteen pain rats and 8 control rats received Rs-fMRI examination to explore the brain functional activity areas, and the regional homogeneity (ReHo) method was used to analyze relevant functional signals among the whole brain. The states of neurons and expression of TRPV1 and NMDRA located in the abnormal ReHo signal brain regions were observed using Nissl staining, qRT-PCR and immunohistochemistry. Results The rats were divided into a pain group and a control group based on the different syndromes and behavioral assessments. We detected significant enhancement of ReHo signals in the anterior cingulate cortex, hippocampus, and thalamus and a reduction in the ReHo values in the basomedial amygdaloid nucleus (BM) and primary motor cortex (M1) in the pain rat group via Rs-fMRI examination. The number of Nissl bodies and apoptotic neurons was increased; moreover, the volume of neurons increased compensatorily in the cingulate cortex, thalamus and hippocampus in the pain group. TRPV1 and NMDRA were overexpressed in apoptotic neurons in the higher ReHo value brain regions in the endometriosis pain group. Conclusion These findings suggest that in rats with endometriosis-associated pain, ReHo signal enhancement was observed in the cingulate cortex, thalamus and hippocampus, which may be due to the increase in the number of apoptotic neurons or the compensatory increase in the volume of overactive neurons.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Shuangzheng Jia
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Dalong Guo
- Air Force Medical Center, PLA, Beijing, People's Republic of China
| | - Sikai Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Wen Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Aoshuang Cheng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Weijie Xie
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, People's Republic of China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, People's Republic of China
| | - Jinhua Leng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
48
|
Effect of endometriosis on the fecal bacteriota composition of mice during the acute phase of lesion formation. PLoS One 2019; 14:e0226835. [PMID: 31887116 PMCID: PMC6936831 DOI: 10.1371/journal.pone.0226835] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence indicates that there is an interaction between the gut microbiota and endometriotic lesions. The new formation of these lesions is associated with stem cell recruitment, angiogenesis and inflammation, which may affect the composition of the gut microbiota. To test this hypothesis, we herein induced endometriotic lesions by transplantation of uterine tissue fragments from green fluorescent protein (GFP)+ donor mice into the peritoneal cavity of GFP- C57BL/6 wild-type mice. Sham-transplanted animals served as controls. Fecal pellets of the animals were collected 3 days before as well as 7 and 21 days after the induction of endometriosis to analyze the composition of the gut microbiota by means of 16S ribosomal RNA gene sequencing. The transplantation of uterine tissue fragments resulted in the establishment of endometriotic lesions in all analyzed mice. These lesions exhibited a typical histomorphology with endometrial glands surrounded by a vascularized stroma. Due to their bright GFP signal, they could be easily differentiated from the surrounding GFP- host tissue. Bacterial 16S rRNA genes were successfully PCR-amplified from the DNA extracts of all obtained mice fecal samples. However, no significant effect of endometriosis induction on the composition of the bacterial microbiota was detected with our experimental setup. Our findings allow careful speculation that endometriosis in mice does not induce pronounced dysbiosis during the acute phase of lesion formation.
Collapse
|
49
|
Positron emission tomography imaging of vascular endothelial growth factor with 64Cu-labeled bevacizumab for non-invasive diagnosis of endometriosis. JOURNAL OF ENDOMETRIOSIS AND PELVIC PAIN DISORDERS 2019. [DOI: 10.1177/2284026519891576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Introduction: Non-invasive diagnosis of endometriosis remains challenging. A promising approach for diagnosing endometriosis is the molecular imaging of vascular endothelial growth factor because angiogenesis plays a role in the establishment of endometriosis. This study aimed to evaluate the potential of copper-64-labeled bevacizumab, an anti–vascular endothelial growth factor antibody, for endometriosis imaging. Methods: Mouse endometriosis model was prepared by autologous transplantation. The vascular endothelial growth factor expression was evaluated by immunohistochemical staining. Biodistribution study and positron emission tomography imaging were performed at 1, 24, and 48 h after the injection of radiolabeled bevacizumab. Results: The immunohistochemical staining revealed that vascular endothelial growth factor is expressed around the stroma and glandular epithelial cells in the endometriosis lesion. The biodistribution study showed a high uptake of indium-111 bevacizumab in the endometriosis lesion. Positron emission tomography imaging with copper-64-labeled bevacizumab clearly visualized the endometriosis lesions at 24 and 48 h after injection. Conclusion: These results indicate the potential usefulness of copper-64-labeled bevacizumab for endometriosis imaging.
Collapse
|
50
|
van Aken MA, Groothuis PG, Panagiotou M, Duin MV, Nap AW, van Rijn TC, Kozicz T, Braat DD, Peeters AB. An objective and automated method for evaluating abdominal hyperalgesia in a rat model for endometriosis. Lab Anim 2019; 54:365-372. [PMID: 31366270 DOI: 10.1177/0023677219856915] [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: 11/17/2022]
Abstract
Chronic pain and subfertility are the main symptoms of concern in women with endometriosis. In order to find new therapeutic options to suppress the pain, translational animal models are indispensable. We have developed a new automated, experimental setup, with full consideration for animal wellbeing, to determine whether operant behaviour can reveal abdominal hyperalgesia in rats with surgically-induced endometriosis, in order to assess whether abdominal hyperalgesia affect behavioural parameters. Endometriosis was induced by transplantation of uterine fragments in the abdominal cavity. Control groups consisted of sham-operated rats and non-operated rats. We have developed an operant chamber (Skinnerbox) which includes a barrier. The rat can climb the barrier in order to reach the food pellet, increasing in this way the pressure to the abdomen. We show that endometriosis rats collect significantly less sugar pellets when compared with the control rats after the introduction of the barrier. In the Skinnerbox experiment, we showed that in a positive operant setting, the introduction of a barrier results in a contrast of operant behaviour of endometriosis rats and control groups, perchance as a result of abdominal discomfort/hyperalgesia due to surgically-induced endometriosis. This is a promising start for the further development of a refined animal model to monitor abdominal discomfort/hyperalgesia in rats with surgically-induced endometriosis.
Collapse
Affiliation(s)
- Mieke Aw van Aken
- Department of Anatomy, Radboud University Medical Centre Nijmegen, The Netherlands.,Department of Gynaecology and Obstetrics, Rijnstate, The Netherlands.,Department of Obstetrics and Gynaecology, Radboud University Medical Centre Nijmegen, The Netherlands
| | | | | | | | - Annemiek W Nap
- Department of Gynaecology and Obstetrics, Rijnstate, The Netherlands
| | - Tineke Cm van Rijn
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, The Netherlands
| | - Tamas Kozicz
- Department of Anatomy, Radboud University Medical Centre Nijmegen, The Netherlands.,Department of Clinical Genomics, Mayo Clinic, USA
| | - Didi Dm Braat
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre Nijmegen, The Netherlands
| | - Ard Bwmm Peeters
- Department of Anatomy, Radboud University Medical Centre Nijmegen, The Netherlands
| |
Collapse
|