1
|
Özbey G, Tuncay G, Düz SA, Çiğremiş Y, Karaer A. The Effect of Endometrial Polyp and Myoma Uteri on Fertility-Related Genes in the Endometrium. Reprod Sci 2025; 32:728-737. [PMID: 39915377 PMCID: PMC11870916 DOI: 10.1007/s43032-025-01802-0] [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: 08/07/2024] [Accepted: 01/22/2025] [Indexed: 03/03/2025]
Abstract
Endometrial polyps are hyperplastic overgrowths of the endometrium that contain both glands and stroma. Myoma uteri is the most common benign tumor of the female pelvis and uterus. HOXA10, which is involved during the organogenesis of the uterus in the embryonic period. The aim of this study was to compare the expression levels of infertility-related genes in endometrial tissue obtained from patients with endometrial polyp and myoma uteri and from healthy controls. A total of 36 patients, including 15 women with endometrial polyp and 21 women with myoma uteri, and 23 healthy controls were enrolled in the study. All patients were evaluated using transvaginal ultrasonography. Endometrial tissue samples were collected from the patient and control groups between the 19th and 21st days of the menstrual cycle. Expression levels of the receptivity markers PROK1, PROKR1, PROK2, PROKR2 and HOXA10 genes were determined by RT- PCR. When the patients diagnosed with endometrial polyp and the healthy controls were compared, it was observed statistically significantly that the expression of PROKR1 increased in endometrium tissue of patients with endometrial polyp (p < 0.05). In patients diagnosed with myoma uteri, gene expression levels of endometrial PROKR1 was statistically significant increased and gene expression levels of PROK1, PROKR2, HOXA10 were found to be statistically significantly decreased compared to the controls (p < 0.05). Changes in the endometrial expression of the HOXA10 and prokineticin gene family in patients with myoma uteri and endometrial polyps may explain certain aspects of infertility in these patients.
Collapse
Affiliation(s)
- Gürkan Özbey
- Department of Obstetrics and Gynecology, Faculty of Medicine, Adıyaman University, Adıyaman, Türkiye.
| | - Görkem Tuncay
- Department of Obstetrics and Gynecology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Senem Arda Düz
- Department of Obstetrics and Gynecology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Yılmaz Çiğremiş
- Department of Medical Biology and Genetics, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Abdullah Karaer
- Department of Obstetrics and Gynecology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| |
Collapse
|
2
|
Grassi A, Rocca MS, Noventa M, Pozzato G, Pozzato A, Scioscia M, Andrisani A, Pontrelli G, Foresta C, De Toni L. In Vitro Gene Expression Profiling of Quantum Molecular Resonance Effects on Human Endometrium Models: A Preliminary Study. Genes (Basel) 2025; 16:290. [PMID: 40149442 PMCID: PMC11942151 DOI: 10.3390/genes16030290] [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: 01/30/2025] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVES The identification of methods to improve the endometrial receptivity (ER) is increasingly of interest. The effect of the electromagnetic field associated with Quantum Molecular Resonance (QMR) on ER was investigated here. METHODS Ishikawa cells were used to evaluate the effects of QMR both on the expression of a group of genes involved in ER, i.e., HOXA10, HOXA11, LIF, ITGB3, and ITGAV, and on cell toxicity. Endometrial samples were obtained from six patients during routine diagnostic procedures, four of which were subsequently used to assess the transcriptional response to QMR through microarray. RESULTS Compared to unexposed controls, a single exposure of Ishikawa cells to QMR for 20 min was associated with a significant and power-dependent up-regulation of all the selected ER-related genes up to 8 power units (PU). Repeated exposure to QMR, up to three consecutive days, showed a significant up-regulation of all the selected genes at power values of 4 PU, from day two onwards. Negligible cytotoxicity was observed. Gene set enrichment analysis, on microarray data of endometrial biopsies stimulated for three consecutive days at 4 PU, showed a significant enrichment of specific gene sets, related to the proteasome system, the cell adhesion, the glucocorticoid receptor, and cell cycle pathways. CONCLUSIONS Our results suggest a possible favorable impact of QMR on ER.
Collapse
Affiliation(s)
- Angela Grassi
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Maria Santa Rocca
- Department of Medicine, University of Padova, 35128 Padova, Italy; (M.S.R.); (C.F.)
| | - Marco Noventa
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padova, 35100 Padova, Italy; (M.N.); (A.A.)
| | | | - Alessandro Pozzato
- Telea Electronic Engineering S.r.l., 36066 Sandrigo, Italy; (G.P.); (A.P.)
| | - Marco Scioscia
- Unit of Gynecological Surgery, Mater Dei Hospital, 70125 Bari, Italy;
| | - Alessandra Andrisani
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padova, 35100 Padova, Italy; (M.N.); (A.A.)
| | - Giovanni Pontrelli
- Department of Obstetrics and Gynecology, Policlinico Hospital, 35031 Abano Terme, Italy;
| | - Carlo Foresta
- Department of Medicine, University of Padova, 35128 Padova, Italy; (M.S.R.); (C.F.)
| | - Luca De Toni
- Department of Medicine, University of Padova, 35128 Padova, Italy; (M.S.R.); (C.F.)
| |
Collapse
|
3
|
Ducreux B, Patrat C, Firmin J, Ferreux L, Chapron C, Marcellin L, Parpex G, Bourdon M, Vaiman D, Santulli P, Fauque P. Systematic review on the DNA methylation role in endometriosis: current evidence and perspectives. Clin Epigenetics 2025; 17:32. [PMID: 39985111 PMCID: PMC11846336 DOI: 10.1186/s13148-025-01828-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/30/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Endometriosis appears to have a multilayered etiology, with genetic and epigenetic factors each contributing half of the pathogenesis. The molecular processes that underlie the onset of endometriosis are yet unclear, but it is assumed that an important contributor in the etiopathology of the disease is DNA methylation. METHODS We conducted a systematic review of the literature regarding DNA methylation in endometriosis following PRISMA guidelines. Records were obtained from PubMed and Web of Science on May 31, 2024. Original research articles analyzing regional or genome-wide DNA methylation in patients with confirmed endometriosis (by surgery and/or histological examination) were given consideration for inclusion. Only human studies were included, and there were no restrictions on the types of tissue that was analyzed (i.e., endometrium, blood, or fetal tissue). The study selection process was run by two manual reviewers. In parallel, an adapted virtual artificial intelligence-powered reviewer operated study selection and results were compared with the manual reviewers' selection. Studies were divided into targeted (e.g., single gene or region level) and epigenome-wide association studies. For each, we extracted a list of genes studied with precise location of CpGs analyzed and the DNA methylation status according to the groups compared. Quality assessment of studies was performed following the Newcastle-Ottawa scale. Quality of evidence was graded following the Grading of Recommendations Assessment, Development and Evaluation. RESULTS A total of 955 studies were screened, and 70 were identified as relevant for systematic review. Our analyses displayed that endometriosis could be polyepigenetic and with alterations in specific genes implicated in major signaling pathways contributing to the disease etiopathology (cell proliferation, differentiation, and division [PI3K-Akt and Wnt-signaling pathway], cell division [MAPK pathway], cell adhesion, cell communication, developmental processes, response to hormone, apoptosis, immunity, neurogenesis, and cancer). CONCLUSION Our systematic review indicates that endometriosis is associated with DNA methylation modifications at specific genes involved in key endometrial biological processes, particularly in the ectopic endometrium. As DNA methylation appears to be an integral component of the pathogenesis of endometriosis, the identification of DNA methylation biomarkers would likely help better understand its causes and aggravating factors as well as potentially facilitate its diagnosis and support the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Bastien Ducreux
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Centre Hospitalier Universitaire (CHU), Faculty of Medicine, INSERM 1231, Université de Bourgogne-Europe, Dijon, France
| | - Catherine Patrat
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Department of Reproductive Biology-CECOS, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Julie Firmin
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Department of Reproductive Biology-CECOS, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Lucile Ferreux
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Department of Reproductive Biology-CECOS, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Charles Chapron
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Department of Gynecology-Obstetric and Assisted Reproduction, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Louis Marcellin
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Department of Gynecology-Obstetric and Assisted Reproduction, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Guillaume Parpex
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Department of Gynecology-Obstetric and Assisted Reproduction, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Mathilde Bourdon
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Department of Gynecology-Obstetric and Assisted Reproduction, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Daniel Vaiman
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
| | - Pietro Santulli
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France
- Department of Gynecology-Obstetric and Assisted Reproduction, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Patricia Fauque
- Faculty of Medicine, Inserm U1016, Université de Paris Cité, 75014, Paris, France.
- Department of Reproductive Biology-CECOS, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France.
| |
Collapse
|
4
|
Retis-Resendiz AM, Gómez-Suárez SK, García-Gómez E, Vázquez-Martínez ER. Molecular Basis of Impaired Decidualization in the Eutopic Endometrium of Endometriosis Patients. Cells 2025; 14:326. [PMID: 40072055 PMCID: PMC11899082 DOI: 10.3390/cells14050326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 03/15/2025] Open
Abstract
Endometriosis is a chronic gynecological disorder characterized by the presence of endometrial tissue outside the uterine cavity. A common feature of this pathology is the impaired decidualization of endometrial stromal cells, a critical process that prepares the uterus for embryo implantation. This decidualization defect has been mechanistically linked to progesterone resistance in endometriotic lesions. However, the presence and underlying mechanisms of decidualization defects in the eutopic endometrium of women with endometriosis remain controversial. The aim of the present study is to integrate and discuss molecular evidence from both in vivo and in vitro studies examining decidualization alterations in the eutopic endometrium of patients with endometriosis. Multiple studies have demonstrated impaired decidualization in the eutopic endometrium of women with endometriosis. These alterations have been reported on multiple genes, signaling pathways, and epigenetic processes. However, additional functional studies are warranted to elucidate whether these decidualization defects directly contribute to endometriosis-associated infertility. A better understanding of the decidualization process and its dysregulation in endometriosis will not only advance the development of targeted fertility treatments but also facilitate the design of more effective therapeutic strategies for managing this chronic condition.
Collapse
Affiliation(s)
- Alejandra Monserrat Retis-Resendiz
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología (INPer)-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 11000, Mexico; (A.M.R.-R.); (S.K.G.-S.)
| | - Sandra Karen Gómez-Suárez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología (INPer)-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 11000, Mexico; (A.M.R.-R.); (S.K.G.-S.)
| | - Elizabeth García-Gómez
- Secretaría de Ciencia, Humanidades, Tecnologías e Innovación (SECIHTI)-Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología (INPer)-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 11000, Mexico;
| | - Edgar Ricardo Vázquez-Martínez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología (INPer)-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 11000, Mexico; (A.M.R.-R.); (S.K.G.-S.)
| |
Collapse
|
5
|
Yin Y, Haller M, Goldinger L, Bharadwaj S, So E, Robles-Pinos V, Chen D, Ma L. Retinoic acid antagonizes estrogen signaling to maintain adult uterine cell fate. Proc Natl Acad Sci U S A 2025; 122:e2416089122. [PMID: 39874292 PMCID: PMC11804538 DOI: 10.1073/pnas.2416089122] [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: 08/08/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
Classical tissue recombination experiments demonstrate that cell-fate determination along the anterior-posterior axis of the Müllerian duct occurs prior to postnatal day 7 in mice. However, little is known about how these cell types are maintained in adults. In this study, we provide genetic evidence that a balance between antagonistic retinoic acid (RA) and estrogen signaling activity is required to maintain simple columnar cell fate in adult uterine epithelium. Transdifferentiation of simple columnar uterine epithelium into stratified cervicovaginal-like epithelium was observed in three related mouse genetic models, in which RA signaling was perturbed in the postnatal uterus. Single-cell RNA sequencing analysis identified the transformed epithelial cell populations and revealed extensive immune cell infiltration resulting from loss of RA signaling. Surprisingly, disruption of RA signaling led to dysregulated expression of a substantial number of estrogen target genes, suggesting that these two pathways may functionally oppose each other in determining and maintaining uterine epithelial cell fate. Consistent with this model, neonatal exposure to the strong synthetic estrogen, diethylstilbestrol, downregulated expression of a group of RA target genes and led to epithelial stratification and immune cell infiltration in wild-type uterus. Treating RA receptor triple conditional knockout pups with fulvestrant, an estrogen antagonist, reestablished the balance between the two signaling pathways, and effectively prevented the transformation of mutant simple columnar epithelia to metaplastic stratified epithelia. These findings implicate an essential role for RA signaling in maintaining uterine cytodifferentiation by antagonizing estrogen signaling in the postnatal uterus.
Collapse
Affiliation(s)
- Yan Yin
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Meade Haller
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Lauren Goldinger
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Shivani Bharadwaj
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Emily So
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Vivian Robles-Pinos
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - David Chen
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Liang Ma
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
6
|
Hou S, Zhang J, Zhang Z, Qu H, Li S, Jiang Y, Liu C. Single-cell transcriptomic atlas of different endometriosis indicating that an interaction between endometriosis-associated mesothelial cells (EAMCs) and ectopic stromal cells may influence progesterone resistance. Clin Transl Med 2025; 15:e70216. [PMID: 39968688 PMCID: PMC11836620 DOI: 10.1002/ctm2.70216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Endometriosis is a hormone-dependent disease, which can usually be divided into peritoneal endometriosis (PEM), deep-infiltrating endometriosis (DIE) and ovarian endometriosis (OEM). Although the three pathologic types are essentially the same disease, they differ in pathological manifestations, molecular features, pain symptoms and hormonal responsiveness. However, there is limited literature focusing on the differences among these types. In this study, we employed single-cell RNA sequencing (scRNA-seq) to profile the transcriptome of each type using surgical biopsy samples obtained from six patients. We aimed to explore and elucidate the variations among these different types of endometriosis. RESULTS We identified five major cell types and 44 subpopulations, including the presence of mesothelial cells in all pathological types, including OEM. Furthermore, we characterised the variations in cell types across different pathological types by employing enrichment analysis to assess functions and pathways. Notably, our findings reveal distinct levels of epithelial-mesenchymal transition (EMT) processes experienced by mesothelial cells within the microenvironment of endometriotic lesions. Through ligand-receptor analysis and referencing relevant literature, we propose that mesothelial cells exert an influence on progesterone resistance in stromal cells through intercellular communication mediated by the FN1-AKT pathway. CONCLUSIONS Our study comprehensively characterises the heterogeneity of the different pathologic types of endometriosis and offers valuable insights into the underlying mechanisms contributing to variations in progesterone resistance across the three subtypes. KEY POINTS Single-cell RNA (ScRNA) atlas across types of endometriosis is established. Mesothelial cells are founded in ovarian endometriosis. Endometriosis-associated mesothelial cells (EAMCs) experience various level of epithelial-mesenchymal transition (EMT) process in different subtypes. EAMCs may exert an influence on progesterone resistance in stromal cells through intercellular communication mediated by the FN1-AKT pathway.
Collapse
Affiliation(s)
- Shengdi Hou
- Department of Obstetrics and GynecologyCapital Medical University Affiliated Beijing Chaoyang HospitalBeijingChina
| | - Jing Zhang
- Department of Obstetrics and GynecologyCapital Medical University Affiliated Beijing Chaoyang HospitalBeijingChina
| | - Zhiqiang Zhang
- Department of Obstetrics and GynecologyCapital Medical University Affiliated Beijing Chaoyang HospitalBeijingChina
| | - Hong Qu
- Department of Obstetrics and GynecologyCapital Medical University Affiliated Beijing Chaoyang HospitalBeijingChina
| | - Shuhong Li
- Department of Obstetrics and GynecologyCapital Medical University Affiliated Beijing Chaoyang HospitalBeijingChina
| | - Ying Jiang
- Department of Obstetrics and GynecologyCapital Medical University Affiliated Beijing Chaoyang HospitalBeijingChina
| | - Chongdong Liu
- Department of Obstetrics and GynecologyCapital Medical University Affiliated Beijing Chaoyang HospitalBeijingChina
| |
Collapse
|
7
|
Mishra A, Modi D. Role of HOXA10 in pathologies of the endometrium. Rev Endocr Metab Disord 2025; 26:81-96. [PMID: 39499452 DOI: 10.1007/s11154-024-09923-8] [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] [Accepted: 10/24/2024] [Indexed: 11/07/2024]
Abstract
HOXA10 belongs to the homeobox gene family and is essential for uterine biogenesis, endometrial receptivity, embryo implantation, and stromal cell decidualization. Available evidence suggests that the expression of HOXA10 is dysregulated in different endometrial disorders like endometrial hyperplasia, endometrial cancer, adenomyosis, endometriosis, recurrent implantation failure, and unexplained infertility. The downregulation of HOXA10 occurs by genetic changes in the HOXA10 gene, methylation of the HOXA10 locus, or selected miRNAs. Endocrine disruptors and organic pollutants also cause the reduced expression of HOXA10 in these conditions. In vivo experiments in mouse models and in vitro studies in human cell lines demonstrate that downregulation of HOXA10 leads to endometrial epithelial cell proliferation, failure of stromal cell decidualization, altered expression of genes involved in cell cycle regulation, immunomodulation, and various signaling pathways. These disruptions are speculated to cause infertility associated with the disorders of the endometrium.
Collapse
Affiliation(s)
- Anuradha Mishra
- Molecular and Cellular Biology Laboratory, National Institute for Research in Reproductive and Child Health, J.M. Street, Parel, Mumbai, 400 012, India
| | - Deepak Modi
- Molecular and Cellular Biology Laboratory, National Institute for Research in Reproductive and Child Health, J.M. Street, Parel, Mumbai, 400 012, India.
| |
Collapse
|
8
|
Stratopoulou CA, Rossi M, Beaussart C, Zipponi M, Camboni A, Donnez J, Dolmans MM. Generation of epithelial-stromal assembloids as an advanced in vitro model of impaired adenomyosis-related endometrial receptivity. Fertil Steril 2025; 123:350-360. [PMID: 39197515 DOI: 10.1016/j.fertnstert.2024.08.339] [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: 05/23/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024]
Abstract
OBJECTIVE To create a novel, more advanced in vitro model of human endometrium, using so-called assembloids, looking to explore endometrial receptivity in adenomyosis. DESIGN Evaluation of assembloid responsiveness to hormonal stimulation by immunohistochemistry, enzyme-linked immunosorbent assay, and scanning electron microscopy. SUBJECTS Twelve women, six of whom were affected by adenomyosis. EXPOSURE Organoids (in the form of glandular fragments) and stromal fibroblasts were collected from endometrial biopsies. The two populations were combined inside an extracellular matrix to create 3D assembloids, which were then exposed to hormonal stimulation (β-estradiol for 48 hours, followed by β-estradiol/progesterone/cyclic adenosine monophosphate for 72 hours) to mimic the window of implantation. MAIN OUTCOME MEASURE(S) Glycodelin, leukemia inhibitory factor (LIF), and homeobox A10 (HOXA10) expression, prolactin secretion, and pinopode development. RESULT(S) Endometrial organoids and stromal cells were successfully isolated from women with and without adenomyosis and combined to generate the assembloid model. On stimulation, assembloids from both groups acquired a more secretory phase-like phenotype, as demonstrated by histology, and were shown to be positive for glycodelin, LIF, and HOXA10 by immunohistochemistry. Adenomyotic assembloids expressed significantly lower levels of LIF and HOXA10 within the stromal compartment after stimulation than did healthy assembloids in the same condition. Enzyme-linked immunosorbent assay revealed prolactin secretion in vitro, showing an upward trend in hormonally treated assembloids from both healthy and affected women. By scanning electron microscopy, fully formed pinopodes were discerned on the epithelial surface of healthy assembloids after stimulation, but they were absent in case of adenomyosis. CONCLUSION(S) Primary assembloids can be generated from endometrial biopsies from both healthy subjects and women affected by adenomyosis. These assembloids are amenable to hormonal stimulation and mimic secretory phase-specific characteristics of endometrial tissue in vivo, including glycodelin, LIF, and HOXA10 expression, and pinopode formation. Assembloids from adenomyosis appear to be less sensitive to hormonal treatment, showing reduced expression of LIF and HOXA10 in the stromal compartment and failing to form pinopodes. All in all, endometrial assembloids may serve as an advanced preclinical model of adenomyosis-related impaired endometrial receptivity, opening up new horizons in understanding and treating the condition.
Collapse
Affiliation(s)
- Christina Anna Stratopoulou
- Gynecology Research Unit, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Margherita Rossi
- Gynecology Research Unit, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium; Obstetrics and Gynecology, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Careggi University Hospital, Florence, Italy
| | - Chloé Beaussart
- Gynecology Research Unit, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Margherita Zipponi
- Gynecology Research Unit, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Camboni
- Gynecology Research Unit, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium; Anatomopathology Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Jacques Donnez
- Société de Recherche pour l'Infertilité, Brussels, Belgium; Université Catholique de Louvain, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium; Gynecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
9
|
Kopec K, Quaranto D, DeSouza NR, Jarboe T, Islam HK, Moscatello A, Li XM, Geliebter J, Tiwari RK. The HOX Gene Family's Role as Prognostic and Diagnostic Biomarkers in Hematological and Solid Tumors. Cancers (Basel) 2025; 17:262. [PMID: 39858044 PMCID: PMC11763641 DOI: 10.3390/cancers17020262] [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: 12/11/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
The HOX gene family encodes for regulatory transcription factors that play a crucial role in embryogenesis and differentiation of adult cells. This highly conserved family of genes consists of thirty-nine genes in humans that are located in four clusters, A-D, on different chromosomes. While early studies on the HOX gene family have been focused on embryonic development and its related disorders, research has shifted to examine aberrant expression of HOX genes and the subsequent implication in cancer prediction and progression. Due to their role of encoding master regulatory transcription factors, the abnormal expression of HOX genes has been shown to affect all stages of tumorigenesis and metastasis. This review highlights the novel role of the HOX family's clinical relevance as both prognostic and diagnostic biomarkers in hematological and solid tumors.
Collapse
Affiliation(s)
- Kaci Kopec
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
| | - Danielle Quaranto
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
| | - Nicole R. DeSouza
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
| | - Tara Jarboe
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
| | - Humayun K. Islam
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| | - Augustine Moscatello
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| | - Xiu-Min Li
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
- Department of Dermatology, New York Medical College, Valhalla, NY 10595, USA
| | - Jan Geliebter
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| | - Raj K. Tiwari
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA; (K.K.); (D.Q.); (N.R.D.); (T.J.); (H.K.I.); (A.M.); (X.-M.L.); (R.K.T.)
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
10
|
Pallavi S, Jain S, Mohanty SK, Andrabi SW, Rajender S. Retinoic Acid Regulates Spermiogenesis Via Hoxb1 and Shh Signaling in Testicular Germ Cells. Reprod Sci 2024; 31:3400-3412. [PMID: 39080234 DOI: 10.1007/s43032-024-01648-y] [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: 04/05/2024] [Accepted: 07/08/2024] [Indexed: 11/02/2024]
Abstract
Retinoic acid (RA) regulates all four major events in spermatogenesis; spermatogonial differentiation, meiotic entry, spermiogenesis, and spermiation. For the pre-meiotic phase, Sertoli cells are the source of RA and for the post-meiotic phase, pachytene spermatocytes are the source of RA. While the entire spermatogenic process is regulated by RA, how each of these phases is regulated by RA remains completely unknown. Homeobox B1 (Hoxb1) has two retinoic acid response elements (RARE) upstream and downstream of the gene. In this study, we investigated if RA facilitates spermatogenesis by its action on Hoxb1. The expressions of the Hoxb1 and Sonic hedgehog (Shh) genes were analyzed in the post-natal mouse testes and the testicular localizations of Hoxb1, Shh and Gli1 were analyzed by immunohistochemistry in the adult rat testis. To delineate the signaling mechanisms, Hoxb1 expression was altered in vitro and in vivo using retinoic acid and miR-361-3p. Finally, the levels of miR-361-3p and HOXB1 were analyzed in infertile human sperm samples. Hoxb1 and Shh gene expressions were found to be low in the testis of post-natal Swiss mice of 7, 14, 28, 35, and 60 days, after which the expressions of both spiked. Immunohistochemistry in the adult mouse testis showed the expressions of Hoxb1, Shh, and Gli1 in the elongating spermatids. Exposure of GC2 cells to RA and in vivo IP RA injection upregulated Hoxb1 and Shh signaling in the testis with increased expressions of Shh, Gli1, and Hdac1. Retinoic acid administration in Swiss mice compromised sperm production and reduced epididymal sperm count. The analysis of infertile human semen samples revealed an increased level of HOXB1 and a decreased level of miR-361-3p as compared to fertile controls. We conclude that retinoic acid regulates late stage of spermatogenesis (spermiogenesis) by affecting Hoxb1 and Shh signaling.
Collapse
Affiliation(s)
- Saini Pallavi
- Division of Endocrinology, Central Drug Research Institute, Uttar Pradesh, Lucknow, India
- Academy of Scientific and Innovative Research, Uttar Pradesh, Ghaziabad, India
| | - Simran Jain
- Division of Endocrinology, Central Drug Research Institute, Uttar Pradesh, Lucknow, India
| | - Sujit Kumar Mohanty
- Division of Endocrinology, Central Drug Research Institute, Uttar Pradesh, Lucknow, India
| | | | - Singh Rajender
- Division of Endocrinology, Central Drug Research Institute, Uttar Pradesh, Lucknow, India.
- Academy of Scientific and Innovative Research, Uttar Pradesh, Ghaziabad, India.
| |
Collapse
|
11
|
Shenoy US, Adiga D, Alhedyan F, Kabekkodu SP, Radhakrishnan R. HOXA9 transcription factor is a double-edged sword: from development to cancer progression. Cancer Metastasis Rev 2024; 43:709-728. [PMID: 38062297 PMCID: PMC11156722 DOI: 10.1007/s10555-023-10159-2] [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: 08/09/2023] [Accepted: 11/30/2023] [Indexed: 04/02/2024]
Abstract
The HOXA9 transcription factor serves as a molecular orchestrator in cancer stemness, epithelial-mesenchymal transition (EMT), metastasis, and generation of the tumor microenvironment in hematological and solid malignancies. However, the multiple modes of regulation, multifaceted functions, and context-dependent interactions responsible for the dual role of HOXA9 as an oncogene or tumor suppressor in cancer remain obscure. Hence, unravelling its molecular complexities, binding partners, and interacting signaling molecules enables us to comprehend HOXA9-mediated transcriptional programs and molecular crosstalk. However, it is imperative to understand its central role in fundamental biological processes such as embryogenesis, foetus implantation, hematopoiesis, endothelial cell proliferation, and tissue homeostasis before designing targeted therapies. Indeed, it presents an enormous challenge for clinicians to selectively target its oncogenic functions or restore tumor-suppressive role without altering normal cellular functions. In addition to its implications in cancer, the present review also focuses on the clinical applications of HOXA9 in recurrence and drug resistance, which may provide a broader understanding beyond oncology, open new avenues for clinicians for accurate diagnoses, and develop personalized treatment strategies. Furthermore, we have also discussed the existing therapeutic options and accompanying challenges in HOXA9-targeted therapies in different cancer types.
Collapse
Affiliation(s)
- U Sangeetha Shenoy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Faisal Alhedyan
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
12
|
Jia S, Zhao F. Single-cell transcriptomic profiling of the neonatal oviduct and uterus reveals new insights into upper Müllerian duct regionalization. FASEB J 2024; 38:e23632. [PMID: 38686936 PMCID: PMC11095678 DOI: 10.1096/fj.202400303r] [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: 02/07/2024] [Revised: 03/20/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024]
Abstract
The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper MD development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.
Collapse
Affiliation(s)
- Shuai Jia
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Fei Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
13
|
Wellik DM. Hox genes and patterning the vertebrate body. Curr Top Dev Biol 2024; 159:1-27. [PMID: 38729674 DOI: 10.1016/bs.ctdb.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The diversity of vertebrate body plans is dizzying, yet stunning for the many things they have in common. Vertebrates have inhabited virtually every part of the earth from its coldest to warmest climates. They locomote by swimming, flying, walking, slithering, or climbing, or combinations of these behaviors. And they exist in many different sizes, from the smallest of frogs, fish and lizards to giraffes, elephants, and blue whales. Despite these differences, vertebrates follow a remarkably similar blueprint for the establishment of their body plan. Within the relatively small amount of time required to complete gastrulation, the process through which the three germ layers, ectoderm, mesoderm, and endoderm are created, the embryo also generates its body axis and is simultaneously patterned. For the length of this axis, the genes that distinguish the neck from the rib cage or the trunk from the sacrum are the Hox genes. In vertebrates, there was evolutionary pressure to maintain this set of genes in the organism. Over the past decades, much has been learned regarding the regulatory mechanisms that ensure the appropriate expression of these genes along the main body axes. Genetic functions continue to be explored though much has been learned. Much less has been discerned on the identity of co-factors used by Hox proteins for the specificity of transcriptional regulation or what downstream targets and pathways are critical for patterning events, though there are notable exceptions. Current work in the field is demonstrating that Hox genes continue to function in many organs long after directing early patterning events. It is hopeful continued research will shed light on remaining questions regarding mechanisms used by this important and conserved set of transcriptional regulators.
Collapse
Affiliation(s)
- Deneen M Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States.
| |
Collapse
|
14
|
Kapper C, Oppelt P, Ganhör C, Gyunesh AA, Arbeithuber B, Stelzl P, Rezk-Füreder M. Minerals and the Menstrual Cycle: Impacts on Ovulation and Endometrial Health. Nutrients 2024; 16:1008. [PMID: 38613041 PMCID: PMC11013220 DOI: 10.3390/nu16071008] [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: 02/26/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The role of minerals in female fertility, particularly in relation to the menstrual cycle, presents a complex area of study that underscores the interplay between nutrition and reproductive health. This narrative review aims to elucidate the impacts of minerals on key aspects of the reproductive system: hormonal regulation, ovarian function and ovulation, endometrial health, and oxidative stress. Despite the attention given to specific micronutrients in relation to reproductive disorders, there is a noticeable absence of a comprehensive review focusing on the impact of minerals throughout the menstrual cycle on female fertility. This narrative review aims to address this gap by examining the influence of minerals on reproductive health. Each mineral's contribution is explored in detail to provide a clearer picture of its importance in supporting female fertility. This comprehensive analysis not only enhances our knowledge of reproductive health but also offers clinicians valuable insights into potential therapeutic strategies and the recommended intake of minerals to promote female reproductive well-being, considering the menstrual cycle. This review stands as the first to offer such a detailed examination of minerals in the context of the menstrual cycle, aiming to elevate the understanding of their critical role in female fertility and reproductive health.
Collapse
Affiliation(s)
- Celine Kapper
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Peter Oppelt
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
- Department for Gynaecology, Obstetrics and Gynaecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Clara Ganhör
- Division of Pathophysiology, Institute of Physiology and Pathophysiology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- Clinical Research Institute for Cardiovascular and Metabolic Diseases, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Ayberk Alp Gyunesh
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Barbara Arbeithuber
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Patrick Stelzl
- Department for Gynaecology, Obstetrics and Gynaecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Marlene Rezk-Füreder
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| |
Collapse
|
15
|
Mikkelsen TF, Vera-Rodriguez M, Greggains G, Fedorcsák P, Hald K. Effect of endometrial biopsy method on ribonucleic acid quality and gene expression analysis in patients with leiomyoma. F S Rep 2024; 5:72-79. [PMID: 38524201 PMCID: PMC10958711 DOI: 10.1016/j.xfre.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 03/26/2024] Open
Abstract
Objective To compare ribonucleic acid (RNA) quantity and purity in tissue collected with different endometrial sampling methods to establish the optimal tool for use in endometrial gene expression studies. Design Observational study. Setting University hospital. Patients Fourteen patients with submucosal leiomyomas. Interventions Unguided biopsies were obtained using a low-pressure suction device before hysteroscopy from 14 patients with submucosal leiomyomas followed by guided biopsy with a resectoscope loop. Fifty-seven samples were collected: 25 obtained using a suction device and 32 with a loop. Main Outcome Measures Total biopsy weight, RNA purity, and RNA yield for each collection method. After complementary deoxyribonucleic acid synthesis, HOXA10 expression was measured by quantitative polymerase chain reaction in the endometrium overlying and remote from the leiomyoma, as similar expression throughout the cavity was a prerequisite for the use of unguided biopsy method. Results The median weight of the samples was significantly larger when obtained with the low-pressure suction device than with the resectoscope loop (153 vs. 20 mg). The RNA yield was similar (suction curette, 1,625 ng/mg; resectoscope loop, 1,779 ng/mg). The A260-to-A280 ratio was satisfactory for 94.7 % of the samples, with no difference between the groups. The endometrial expression of HOXA10 was similar in areas overlying the leiomyoma compared with that in remote endometrial sites (2-ΔCt = 0.0224 vs. 0.0225). Conclusions Low-pressure endometrial suction devices provide tissue samples with acceptable RNA purity and quantity for gene expression studies. The expression of HOXA10 did not differ between endometrial sampling sites even in the presence of leiomyomas.
Collapse
Affiliation(s)
- Thea Falkenberg Mikkelsen
- Department of Gynecology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Maria Vera-Rodriguez
- Department of Reproductive Medicine, Oslo University Hospital, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Gareth Greggains
- Department of Reproductive Medicine, Oslo University Hospital, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Péter Fedorcsák
- Department of Reproductive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kirsten Hald
- Department of Gynecology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
16
|
Cormier SA, Kappen C. Identification of a Chondrocyte-Specific Enhancer in the Hoxc8 Gene. J Dev Biol 2024; 12:5. [PMID: 38390956 PMCID: PMC10885077 DOI: 10.3390/jdb12010005] [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: 12/04/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Hox genes encode transcription factors whose roles in patterning animal body plans during embryonic development are well-documented. Multiple studies demonstrate that Hox genes continue to act in adult cells, in normal differentiation, in regenerative processes, and, with abnormal expression, in diverse types of cancers. However, surprisingly little is known about the regulatory mechanisms that govern Hox gene expression in specific cell types, as they differentiate during late embryonic development, and in the adult organism. The murine Hoxc8 gene determines the identity of multiple skeletal elements in the lower thoracic and lumbar region and continues to play a role in the proliferation and differentiation of cells in cartilage as the skeleton matures. This study was undertaken to identify regulatory elements in the Hoxc8 gene that control transcriptional activity, specifically in cartilage-producing chondrocytes. We report that an enhancer comprising two 416 and 224 bps long interacting DNA elements produces reporter gene activity when assayed on a heterologous transcriptional promoter in transgenic mice. This enhancer is distinct in spatial, temporal, and molecular regulation from previously identified regulatory sequences in the Hoxc8 gene that control its expression in early development. The identification of a tissue-specific Hox gene regulatory element now allows mechanistic investigations into Hox transcription factor expression and function in differentiating cell types and adult tissues and to specifically target these cells during repair processes and regeneration.
Collapse
Affiliation(s)
- Stephania A. Cormier
- Department of Respiratory Immunology and Toxicology, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA;
| | - Claudia Kappen
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| |
Collapse
|
17
|
Jia S, Zhao F. Single-cell transcriptomic profiling of the neonatal oviduct and uterus reveals new insights into upper Müllerian duct regionalization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572607. [PMID: 38187777 PMCID: PMC10769252 DOI: 10.1101/2023.12.20.572607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper Müllerian duct development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.
Collapse
|
18
|
Lofrano-Porto A, Pereira SA, Dauber A, Bloom JC, Fontes AN, Asimow N, de Moraes OL, Araujo PAT, Abreu AP, Guo MH, De Oliveira SF, Liu H, Lee C, Kuohung W, Coelho MS, Carroll RS, Jiang R, Kaiser UB. OSR1 disruption contributes to uterine factor infertility via impaired Müllerian duct development and endometrial receptivity. J Clin Invest 2023; 133:e161701. [PMID: 37847567 PMCID: PMC10688984 DOI: 10.1172/jci161701] [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: 05/13/2022] [Accepted: 09/28/2023] [Indexed: 10/18/2023] Open
Abstract
Three sisters, born from consanguineous parents, manifested a unique Müllerian anomaly characterized by uterine hypoplasia with thin estrogen-unresponsive endometrium and primary amenorrhea, but with spontaneous tubal pregnancies. Through whole-exome sequencing followed by comprehensive genetic analysis, a missense variant was identified in the OSR1 gene. We therefore investigated OSR1/OSR1 expression in postpubertal human uteri, and the prenatal and postnatal expression pattern of Osr1/Osr1 in murine developing Müllerian ducts (MDs) and endometrium, respectively. We then investigated whether Osr1 deletion would affect MD development, using WT and genetically engineered mice. Human uterine OSR1/OSR1 expression was found primarily in the endometrium. Mouse Osr1 was expressed prenatally in MDs and Wolffian ducts (WDs), from rostral to caudal segments, in E13.5 embryos. MDs and WDs were absent on the left side and MDs were rostrally truncated on the right side of E13.5 Osr1-/- embryos. Postnatally, Osr1 was expressed in mouse uteri throughout their lifespan, peaking at postnatal days 14 and 28. Osr1 protein was present primarily in uterine luminal and glandular epithelial cells and in the epithelial cells of mouse oviducts. Through this translational approach, we demonstrated that OSR1 in humans and mice is important for MD development and endometrial receptivity and may be implicated in uterine factor infertility.
Collapse
Affiliation(s)
- Adriana Lofrano-Porto
- Molecular Pharmacology Laboratory (FARMOL), Faculty of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
- Section of Endocrinology, Gonadal and Adrenal Diseases Clinics, University Hospital of Brasilia, Brasilia-DF, Brazil
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sidney Alcântara Pereira
- Molecular Pharmacology Laboratory (FARMOL), Faculty of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew Dauber
- Division of Endocrinology, Children’s National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, DC, USA
| | - Jordana C.B. Bloom
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Audrey N. Fontes
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Naomi Asimow
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Olívia Laquis de Moraes
- Molecular Pharmacology Laboratory (FARMOL), Faculty of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Petra Ariadne T. Araujo
- Molecular Pharmacology Laboratory (FARMOL), Faculty of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Ana Paula Abreu
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael H. Guo
- Division of Endocrinology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Silviene F. De Oliveira
- Department of Genetics and Morphology, Institute of Biology, University of Brasilia, Brasilia-DF, Brazil
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Han Liu
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Charles Lee
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Wendy Kuohung
- Department of Obstetrics and Gynecology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Michella S. Coelho
- Molecular Pharmacology Laboratory (FARMOL), Faculty of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Rona S. Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ursula B. Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Jackson R, Rajadhyaksha EV, Loeffler RS, Flores CE, Van Doorslaer K. Characterization of 3D organotypic epithelial tissues reveals tonsil-specific differences in tonic interferon signaling. PLoS One 2023; 18:e0292368. [PMID: 37792852 PMCID: PMC10550192 DOI: 10.1371/journal.pone.0292368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023] Open
Abstract
Three-dimensional (3D) culturing techniques can recapitulate the stratified nature of multicellular epithelial tissues. Organotypic 3D epithelial tissue culture methods have several applications, including the study of tissue development and function, drug discovery and toxicity testing, host-pathogen interactions, and the development of tissue-engineered constructs for use in regenerative medicine. We grew 3D organotypic epithelial tissues from foreskin, cervix, and tonsil-derived primary cells and characterized the transcriptome of these in vitro tissue equivalents. Using the same 3D culturing method, all three tissues yielded stratified squamous epithelium, validated histologically using basal and superficial epithelial cell markers. The goal of this study was to use RNA-seq to compare gene expression patterns in these three types of epithelial tissues to gain a better understanding of the molecular mechanisms underlying their function and identify potential therapeutic targets for various diseases. Functional profiling by over-representation and gene set enrichment analysis revealed tissue-specific differences: i.e., cutaneous homeostasis and lipid metabolism in foreskin, extracellular matrix remodeling in cervix, and baseline innate immune differences in tonsil. Specifically, tonsillar epithelia may play an active role in shaping the immune microenvironment of the tonsil balancing inflammation and immune responses in the face of constant exposure to microbial insults. Overall, these data serve as a resource, with gene sets made available for the research community to explore, and as a foundation for understanding the epithelial heterogeneity and how it may impact their in vitro use. An online resource is available to investigate these data (https://viz.datascience.arizona.edu/3DEpiEx/).
Collapse
Affiliation(s)
- Robert Jackson
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Esha V. Rajadhyaksha
- College of Medicine and College of Science, University of Arizona, Tucson, Arizona, United States of America
| | - Reid S. Loeffler
- Biosystems Engineering, College of Agriculture and Life Sciences, College of Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Caitlyn E. Flores
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Koenraad Van Doorslaer
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, Cancer Biology Graduate Interdisciplinary Program, Genetics Graduate Interdisciplinary Program, and University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
20
|
Lazim N, Elias MH, Sutaji Z, Abdul Karim AK, Abu MA, Ugusman A, Syafruddin SE, Mokhtar MH, Ahmad MF. Expression of HOXA10 Gene in Women with Endometriosis: A Systematic Review. Int J Mol Sci 2023; 24:12869. [PMID: 37629050 PMCID: PMC10454210 DOI: 10.3390/ijms241612869] [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: 07/12/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
The homeobox A10 (HOXA10) gene is known to be related to endometriosis; however, due to a lack of knowledge/evidence in the pathogenesis of endometriosis, the mechanisms that link HOXA10 to endometriosis still need to be clarified. This review addresses the difference in the expression of the HOXA10 gene in endometriotic women versus non-endometriotic women across populations by country and discusses its influences on women's fertility. An organized search of electronic databases was conducted in Scopus, ScienceDirect, PubMed, and Web of Science. The keywords used were (HOXA10 OR "homeobox A10" OR PL OR HOX1 OR HOX1H OR HOX1.8) AND ("gene expression") AND (endometriosis). The initial search resulted in 623 articles, 10 of which were included in this review. All ten papers included in this study were rated fair in terms of the quality of the studies conducted. The expression of the HOXA10 gene was found to be downregulated in most studies. However, one study provided evidence of the downregulation and upregulation of HOXA10 gene expression due to the localization of endometriotic lesions. Measuring the expression of the HOXA10 gene in women is clinically essential to predicting endometriosis, endometrial receptivity, and the development of pinopodes in the endometrium during the luteal phase.
Collapse
Affiliation(s)
- Nurunnajah Lazim
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (N.L.); (M.H.E.); (Z.S.); (M.A.A.); (A.K.A.K.)
| | - Marjanu Hikmah Elias
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (N.L.); (M.H.E.); (Z.S.); (M.A.A.); (A.K.A.K.)
- Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia, Nilai 71800, Negeri Sembilan, Malaysia
| | - Zulazmi Sutaji
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (N.L.); (M.H.E.); (Z.S.); (M.A.A.); (A.K.A.K.)
- Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia, Nilai 71800, Negeri Sembilan, Malaysia
| | - Abdul Kadir Abdul Karim
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (N.L.); (M.H.E.); (Z.S.); (M.A.A.); (A.K.A.K.)
| | - Mohammad Azrai Abu
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (N.L.); (M.H.E.); (Z.S.); (M.A.A.); (A.K.A.K.)
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (A.U.); (M.H.M.)
| | - Saiful Effendi Syafruddin
- Medical Molecular Biology Institute, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia;
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (A.U.); (M.H.M.)
| | - Mohd Faizal Ahmad
- Advanced Reproductive Centre (ARC) HCTM UKM, Department of Obstetrics & Gynecology, Faculty of Medicine, National University of Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (N.L.); (M.H.E.); (Z.S.); (M.A.A.); (A.K.A.K.)
| |
Collapse
|
21
|
Peinado FM, Olivas-Martínez A, Iribarne-Durán LM, Ubiña A, León J, Vela-Soria F, Fernández-Parra J, Fernández MF, Olea N, Freire C, Ocón-Hernández O, Artacho-Cordón F. Cell cycle, apoptosis, cell differentiation, and lipid metabolism gene expression in endometriotic tissue and exposure to parabens and benzophenones. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 879:163014. [PMID: 37003176 DOI: 10.1016/j.scitotenv.2023.163014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/15/2023] [Accepted: 03/19/2023] [Indexed: 05/17/2023]
Abstract
AIM To describe the expression profile in endometriotic tissue of genes involved in four signaling pathways related to the development and progression of endometriosis (cell cycle, apoptosis, cell differentiation and lipid metabolism) and to explore its relationship with the women exposure to chemicals with hormonal activity released from cosmetics and personal care products (PCPs). METHODS This cross-sectional study, encompassed within the EndEA study, comprised a subsample of 33 women with endometriosis. Expression levels of 13 genes (BMI1, CCNB1, CDK1, BAX, BCL2L1, FOXO3, SPP1, HOXA10, PDGFRA, SOX2, APOE, PLCG1 and PLCG2) in endometriotic tissue and urinary concentrations of 4 paraben (PB) and 3 benzophenone (BP) congeners were quantified. Bivariate linear and logistic regression analyses were performed to explore the associations between exposure and gene expression levels. RESULTS A total of 8 out 13 genes (61.5 %) were expressed in >75 % of the samples. Exposure to congeners of PBs and/or BPs was associated with the overexpression of CDK1 gene (whose protein drives cells through G2 phase and mitosis), HOXA10 and PDGFRA genes (whose proteins favor pluripotent cell differentiation to endometrial cells), and APOE (whose protein regulates the transport and metabolism of cholesterol, triglycerides and phospholipids in multiple tissues) and PLCG2 genes (whose protein creates 1D-myo-inositol 1,4,5-trisphosphate and diacylglycerol, two important second messengers). CONCLUSIONS Our findings suggest that women exposure to cosmetic and PCP-released chemicals might be associated with the promotion of cell cycle and cell differentiation as well as with lipid metabolism disruption in endometriotic tissue, three crucial signaling pathways in the development and progression of endometriosis. However, further studies should be accomplished to confirm these preliminary data.
Collapse
Affiliation(s)
- F M Peinado
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain; University of Granada, Centre for Biomedical Research, E-18016 Granada, Spain; Radiology and Physical Medicine Department, University of Granada, E-18016 Granada, Spain.
| | - A Olivas-Martínez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain; University of Granada, Centre for Biomedical Research, E-18016 Granada, Spain; Radiology and Physical Medicine Department, University of Granada, E-18016 Granada, Spain
| | - L M Iribarne-Durán
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain
| | - A Ubiña
- General surgery, San Cecilio University Hospital, E-18016 Granada, Spain
| | - J León
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain; Digestive Medicine Unit, 'San Cecilio' University Hospital, E-18012 Granada, Spain; CIBER Hepatic and Digestive Diseases (CIBEREHD), E-28029 Madrid, Spain
| | - F Vela-Soria
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain
| | - J Fernández-Parra
- Gynaecology and Obstetrics Unit, 'Virgen de las Nieves' University Hospital, E-18014 Granada, Spain
| | - M F Fernández
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), E-28029 Madrid, Spain; Radiology and Physical Medicine Department, University of Granada, E-18016 Granada, Spain
| | - N Olea
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), E-28029 Madrid, Spain; Radiology and Physical Medicine Department, University of Granada, E-18016 Granada, Spain; Nuclear Medicine Unit, 'San Cecilio' University Hospital, E-18016 Granada, Spain
| | - C Freire
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), E-28029 Madrid, Spain
| | - O Ocón-Hernández
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain; Gynaecology and Obstetrics Unit, 'San Cecilio' University Hospital, E-18016 Granada, Spain
| | - F Artacho-Cordón
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), E-18012 Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), E-28029 Madrid, Spain; Radiology and Physical Medicine Department, University of Granada, E-18016 Granada, Spain.
| |
Collapse
|
22
|
Kałuzińska-Kołat Ż, Kołat D, Kośla K, Płuciennik E, Bednarek AK. Delineating the glioblastoma stemness by genes involved in cytoskeletal rearrangements and metabolic alterations. World J Stem Cells 2023; 15:302-322. [PMID: 37342224 PMCID: PMC10277965 DOI: 10.4252/wjsc.v15.i5.302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/03/2023] [Accepted: 03/08/2023] [Indexed: 05/26/2023] Open
Abstract
Literature data on glioblastoma ongoingly underline the link between metabolism and cancer stemness, the latter is one responsible for potentiating the resistance to treatment, inter alia due to increased invasiveness. In recent years, glioblastoma stemness research has bashfully introduced a key aspect of cytoskeletal rearrangements, whereas the impact of the cytoskeleton on invasiveness is well known. Although non-stem glioblastoma cells are less invasive than glioblastoma stem cells (GSCs), these cells also acquire stemness with greater ease if characterized as invasive cells and not tumor core cells. This suggests that glioblastoma stemness should be further investigated for any phenomena related to the cytoskeleton and metabolism, as they may provide new invasion-related insights. Previously, we proved that interplay between metabolism and cytoskeleton existed in glioblastoma. Despite searching for cytoskeleton-related processes in which the investigated genes might have been involved, not only did we stumble across the relation to metabolism but also reported genes that were found to be implicated in stemness. Thus, dedicated research on these genes in GSCs seems justifiable and might reveal novel directions and/or biomarkers that could be utilized in the future. Herein, we review the previously identified cytoskeleton/metabolism-related genes through the prism of glioblastoma stemness.
Collapse
Affiliation(s)
- Żaneta Kałuzińska-Kołat
- Department of Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Damian Kołat
- Department of Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Katarzyna Kośla
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Elżbieta Płuciennik
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| |
Collapse
|
23
|
Jackson R, Rajadhyaksha EV, Loeffler RS, Flores CE, Van Doorslaer K. Characterization of 3D organotypic epithelial tissues reveals tonsil-specific differences in tonic interferon signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524743. [PMID: 36711548 PMCID: PMC9882319 DOI: 10.1101/2023.01.19.524743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Three-dimensional (3D) culturing techniques can recapitulate the stratified nature of multicellular epithelial tissues. Organotypic 3D epithelial tissue culture methods have several applications, including the study of tissue development and function, drug discovery and toxicity testing, host-pathogen interactions, and the development of tissue-engineered constructs for use in regenerative medicine. We grew 3D organotypic epithelial tissues from foreskin, cervix, and tonsil-derived primary cells and characterized the transcriptome of these in vitro tissue equivalents. Using the same 3D culturing method, all three tissues yielded stratified squamous epithelium, validated histologically using basal and superficial epithelial cell markers. The goal of this study was to use RNA-seq to compare gene expression patterns in these three types of epithelial tissues to gain a better understanding of the molecular mechanisms underlying their function and identify potential therapeutic targets for various diseases. Functional profiling by over-representation and gene set enrichment analysis revealed tissue-specific differences: i.e. , cutaneous homeostasis and lipid metabolism in foreskin, extracellular matrix remodeling in cervix, and baseline innate immune differences in tonsil. Specifically, tonsillar epithelia may play an active role in shaping the immune microenvironment of the tonsil balancing inflammation and immune responses in the face of constant exposure to microbial insults. Overall, these data serve as a resource, with gene sets made available for the research community to explore, and as a foundation for understanding the epithelial heterogeneity and how it may impact their in vitro use. An online resource is available to investigate these data ( https://viz.datascience.arizona.edu/3DEpiEx/ ).
Collapse
Affiliation(s)
- Robert Jackson
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Esha V Rajadhyaksha
- College of Medicine and College of Science, University of Arizona, Tucson, AZ, USA
| | - Reid S Loeffler
- Biosystems Engineering, College of Agriculture and Life Sciences; College of Engineering, University of Arizona, Tucson, AZ, USA
| | - Caitlyn E Flores
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
| | - Koenraad Van Doorslaer
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Department of Immunobiology; Cancer Biology Graduate Interdisciplinary Program; Genetics Graduate Interdisciplinary Program; and University of Arizona Cancer Center, University of Arizona, Tucson, AZ USA
| |
Collapse
|
24
|
Shanmugam DAS, Dhatchanamurthy S, Leela KA, Bhaskaran RS. Maternal exposure to di(2-ethylhexyl) phthalate (DEHP) causes multigenerational adverse effects on the uterus of F 1 and F 2 offspring rats. Reprod Toxicol 2023; 115:17-28. [PMID: 36435455 DOI: 10.1016/j.reprotox.2022.11.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
Abstract
Phthalates are one of the ubiquitous chemicals found in day-to-day products like food packaging, children's toys, and other consumer commodities. There is rising concern that repeated exposure to phthalates during pregnancy and lactation could have long-term effects on maternal and fetal health. We hypothesize that exposure to DEHP during the developmental windows might affect the expression of molecules that regulate uterine function and that this effect would be passed on to further generations. Rat dams were treated with olive oil (vehicle) or DEHP (100 mg/kg b.wt./day) orally from gestational day 9 (GD 9) to the end of lactation (PND 21). F0 maternal DEHP exposure resulted in multigenerational (F1 and F2) reproductive toxicity, as evidenced by an extended estrous cycle, decreased mating, fertility, and fecundity indices. Serum progesterone and estradiol levels were decreased and their cognate receptors (PR and ERα) in the uterus were decreased in the DEHP-exposed offspring rats. Further analysis of the expression of estrogen and progesterone regulatory genes such as Hox a11, VEGF A, Ihh, LIFR, EP4, PTCH, NR2F2, BMP2, and Wnt4 were reduced in the uteri of adult F1 and F2 generation rats born from DEHP-exposed F0 dams. Decreased expression of these crucial proteins due to DEHP exposure may lead to defects in epithelial proliferation and secretion, uterine receptivity, and decidualization in the uteri of successive generations. This study showed that maternal DEHP exposure impairs the expression of molecules that regulate uterine function and this multigenerational effect is transmitted via maternal lineage.
Collapse
Affiliation(s)
- Dharani Abirama Sundari Shanmugam
- Department of Endocrinology, Dr. ALM. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600113, India
| | - Sakthivel Dhatchanamurthy
- Department of Endocrinology, Dr. ALM. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600113, India
| | - Kamakshi Arjunan Leela
- Department of Endocrinology, Dr. ALM. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600113, India
| | - Ravi Sankar Bhaskaran
- Department of Endocrinology, Dr. ALM. Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600113, India.
| |
Collapse
|
25
|
Abstract
Hox genes encode evolutionarily conserved transcription factors that are essential for the proper development of bilaterian organisms. Hox genes are unique because they are spatially and temporally regulated during development in a manner that is dictated by their tightly linked genomic organization. Although their genetic function during embryonic development has been interrogated, less is known about how these transcription factors regulate downstream genes to direct morphogenetic events. Moreover, the continued expression and function of Hox genes at postnatal and adult stages highlights crucial roles for these genes throughout the life of an organism. Here, we provide an overview of Hox genes, highlighting their evolutionary history, their unique genomic organization and how this impacts the regulation of their expression, what is known about their protein structure, and their deployment in development and beyond.
Collapse
Affiliation(s)
- Katharine A. Hubert
- Program in Genetics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Deneen M. Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
26
|
Bi-potential hPSC-derived Müllerian duct-like cells for full-thickness and functional endometrium regeneration. NPJ Regen Med 2022; 7:68. [DOI: 10.1038/s41536-022-00263-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
AbstractStem cell-based tissue regeneration strategies are promising treatments for severe endometrial injuries. However, there are few appropriate seed cells for regenerating a full-thickness endometrium, which mainly consists of epithelia and stroma. Müllerian ducts in female embryonic development develop into endometrial epithelia and stroma. Hence, we first generated human pluripotent stem cells (hPSC)-derived Müllerian duct-like cells (MDLCs) using a defined and effective protocol. The MDLCs are bi-potent, can gradually differentiate into endometrial epithelial and stromal cells, and reconstitute full-thickness endometrium in vitro and in vivo. Furthermore, MDLCs showed the in situ repair capabilities of reconstructing endometrial structure and recovering pregnancy function in full-thickness endometrial injury rats, and their differentiation fate was revealed by single-cell RNA sequencing (scRNA-seq). Our study provides a strategy for hPSC differentiation into endometrial lineages and an alternative seed cell for injured endometrial regeneration.
Collapse
|
27
|
Qi Y, Wang X, Hou S, Wu Z, Xu X, Pang C. Intracavitary physiotherapy combined with acupuncture mediated AMPK/mTOR signalling to improve endometrial receptivity in patients with thin endometrium. Eur J Obstet Gynecol Reprod Biol 2022; 277:32-41. [DOI: 10.1016/j.ejogrb.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/19/2022] [Accepted: 08/02/2022] [Indexed: 11/04/2022]
|
28
|
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: 7] [Impact Index Per Article: 2.3] [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
|
29
|
Ekanayake DL, Małopolska MM, Schwarz T, Tuz R, Bartlewski PM. The roles and expression of HOXA/Hoxa10 gene: A prospective marker of mammalian female fertility? Reprod Biol 2022; 22:100647. [PMID: 35550944 DOI: 10.1016/j.repbio.2022.100647] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
This review addresses the influence of homebox A10/a10 (HOXA/Hoxa10) gene on reproductive tract anatomy and functional fertility in mammalian species, and discusses major endocrine and environmental regulators of HOXA/Hoxa10 expression. Female reproductive efficiency or success is a function of several factors including the ovulation and fertilization rate, and uterine receptivity. A family of HOX/Hox genes establishes the segmental identity of the reproductive tract during embryogenesis and retains its physiological plasticity in sexually mature animals and humans. In particular, the HOXA/Hoxa10 gene is an intrinsic component of implantation, decidualization, and immunomodulation in the adult uterus. It was, therefore, suggested that knowledge of HOXA/Hoxa10 regulation might be essential in navigating molecular mechanisms with the aim of enhancing female reproductive potential. However, a recent study in pigs revealed a lack of associations between endometrial HOXA10 expression and reproductive tract morphology, and very poor correlations with sows' fertility metrics. Retinoic acid mainly regulates 3' HOX/Hox paralogs but may also modify the expression of downstream HOX/Hox genes, including HOXA/Hoxa10. Sex steroids directly regulate HOXA/Hoxa10 expression. The vitamin D receptor pathway modulates HOXA/Hoxa10 expression in the adult reproductive tract. Lastly, endocrine disruptors such as diethylstilbestrol, methoxychlor, bisphenol A, and isoflavones were shown to alter HOXA/Hoxa10 expression, thus affecting reproductive competence of the female.
Collapse
Affiliation(s)
- Don Lochana Ekanayake
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road, Guelph, Ontario N1G 2W1 Canada
| | - Martyna M Małopolska
- Department of Pig Breeding, National Research Institute of Animal Production, 32-083 Balice n. Kraków, Poland.
| | - Tomasz Schwarz
- Department of Animal Genetics, Breeding and Ethology, Faculty of Animal Science, University of Agriculture in Kraków, 24/28 Mickiewicza Avenue, 31-120 Kraków, Poland
| | - Ryszard Tuz
- Department of Animal Genetics, Breeding and Ethology, Faculty of Animal Science, University of Agriculture in Kraków, 24/28 Mickiewicza Avenue, 31-120 Kraków, Poland
| | - Pawel M Bartlewski
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road, Guelph, Ontario N1G 2W1 Canada
| |
Collapse
|
30
|
Uterine HOXA11 antisense long non-coding RNA prevents decidualization: A new pathway-regulating pregnancy. Mol Ther 2022; 30:1357-1358. [PMID: 35320748 PMCID: PMC9077366 DOI: 10.1016/j.ymthe.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 11/24/2022] Open
|
31
|
Incidence Of Congenital Uterine Abnormalities In Polycystic Ovarian Syndrome. Eur J Obstet Gynecol Reprod Biol 2022; 271:183-188. [DOI: 10.1016/j.ejogrb.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/23/2021] [Accepted: 02/06/2022] [Indexed: 11/18/2022]
|
32
|
Allen-Brady K, Bortolini MAT, Damaser MS. Mouse Knockout Models for Pelvic Organ Prolapse: a Systematic Review. Int Urogynecol J 2022; 33:1765-1788. [PMID: 35088092 DOI: 10.1007/s00192-021-05066-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
INTRODUCTION AND HYPOTHESIS Mouse knockout (KO) models of pelvic organ prolapse (POP) have contributed mechanistic evidence for the role of connective tissue defects, specifically impaired elastic matrix remodeling. Our objective was to summarize what mouse KO models for POP are available and what have we learned from these mouse models about the pathophysiological mechanisms of POP development. METHODS We conducted a systematic review and reported narrative findings according to PRISMA guidelines. Two independent reviewers searched PubMed, Scopus and Embase for relevant manuscripts and conference abstracts for the time frame of January 1, 2000, to March 31, 2021. Conference abstracts were limited to the past 5 years. RESULTS The search strategy resulted in 294 total titles. We ultimately included 25 articles and an additional 11 conference abstracts. Five KO models have been studied: Loxl1, Fbln5, Fbln3, Hoxa11 and Upii-sv40t. Loxl1 and Fbln5 KO models have provided the most reliable and predictable POP phenotype. Loxl1 KO mice develop POP primarily from failure to heal after giving birth, whereas Fbln5 KO mice develop POP with aging. These mouse KO models have been used for a wide variety of investigations including genetic pathways involved in development of POP, biomechanical properties of the pelvic floor, elastic fiber deposition, POP therapies and the pathophysiology associated with mesh complications. CONCLUSIONS Mouse KO models have proved to be a valuable tool in the study of specific genes and their role in the development and progression of POP. They may be useful to study POP treatments and POP complications.
Collapse
Affiliation(s)
- Kristina Allen-Brady
- Department of Internal Medicine, University of Utah, Williams Building 295 Chipeta Way, Salt Lake City, UT, USA.
| | - Maria A T Bortolini
- Department of Gynecology, Sector of Urogynecology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Margot S Damaser
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| |
Collapse
|
33
|
Bi Y, Huang W, Yuan L, Chen S, Liao S, Fu X, Liu B, Yang Y. HOXA10 improves endometrial receptivity by upregulating E-cadherin. Biol Reprod 2022; 106:992-999. [PMID: 35044439 DOI: 10.1093/biolre/ioac007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
In the endometrium of women with recurrent implantation failure (RIF) and unexplained recurrent miscarriage (RM),the expression levels of homeobox A10 (HOXA10) and E-cadherin were positively correlated. To explore whether HOXA10 regulates E-cadherin during endometrial receptivity establishment, Ishikawa and RL95–2 cells were transfected with target-specific siRNA and overexpression plasmid of HOXA10. The expression levels of HOXA10 and E-cadherin were measured by western blot and qRT-PCR. Attachment assay of JEG-3 spheroids to endometrial cells were conducted to explore the adhesive functions after HOXA10 interfered. Chromatin immunoprecipitation assays and dual luciferase reporter were used to investigate the regulatory mechanism of HOXA10. CD1 mice were transfected with si-HOXA10 to confirm these results in vivo. In Ishikawa and RL95–2 cells, the expression of E-cadherin was positively correlated with HOXA10 when it was silenced/overexpressed. Consistently, the adhesion of endometrial epithelium cells and trophoblast cells was inhibited after HOXA10 was silenced, and exogenous restoration of E-cadherin expression reversed this effect to some extent. HOXA10 regulates the expression of E-cadherin by directly binding to a conserved motif (TGTACTAAAAA) located in the E-cadherin promoter region. In addition, after knockdown of HOXA10 in CD1 mice, both the implantation and live birth rates were decreased. In conclusion, HOXA10 can bind to the E-cadherin promoter region and directly regulate its expression, thereby improving endometrial receptivity and subsequently increasing the embryo adhesion and implantation.
Collapse
Affiliation(s)
- Yin Bi
- Reproductive Medicine Centre, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Weiyu Huang
- Reproductive Medicine Centre, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Lifang Yuan
- Reproductive Medicine Centre, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Saiqiong Chen
- Reproductive Medicine Centre, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Shengbin Liao
- Reproductive Medicine Centre, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xiaoqian Fu
- Reproductive Medicine Centre, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Bo Liu
- Reproductive Medicine Centre, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Yihua Yang
- Reproductive Medicine Centre, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| |
Collapse
|
34
|
Zhao H, Hu S, Qi J, Wang Y, Ding Y, Zhu Q, He Y, Lu Y, Yao Y, Wang S, Du Y, Sun Y. Increased expression of HOXA11-AS attenuates endometrial decidualization in recurrent implantation failure patients. Mol Ther 2022; 30:1706-1720. [PMID: 35114391 PMCID: PMC9077377 DOI: 10.1016/j.ymthe.2022.01.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/29/2021] [Accepted: 01/27/2022] [Indexed: 11/19/2022] Open
Abstract
Endometrial decidualization is a prerequisite for implantation, and impaired decidualization is associated with recurrent implantation failure (RIF). Coding genes of the HOX family have been clarified as critical regulators in endometrial decidualization, but the role of long non-coding RNAs (lncRNAs) in the HOX gene family has yet to be determined. The aim of this study was to clarify the possible roles of lncRNAs in the HOX gene family in decidualization. In this study, we identified that HOXA11-AS was the most reduced lncRNA in the HOX family in the human endometrium during the window of implantation, and it was elevated in RIF patients. Mechanistically, HOXA11-AS negatively regulated decidualization through competitive interaction with PTBP1, an RNA-binding protein. Binding of PTBP1 to HOXA11-AS limited PTBP1 availability to regulate PKM1/2 alternative splicing, resulting in enhanced PKM1 and diminished PKM2 expression, thus attenuating decidualization. The pattern of high HOXA11-AS expression and impaired PKM2 splicing was consistently observed in RIF patients. Collectively, our study indicates that the increase of HOXA11-AS is detrimental to endometrial decidualization, likely contributing to RIF. Our study may shed light on the pathogenesis and treatment of RIF.
Collapse
Affiliation(s)
- Hanting Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Shuanggang Hu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jia Qi
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yuan Wang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Ying Ding
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Qinling Zhu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yaqiong He
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yao Lu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yue Yao
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Shiyao Wang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 845 Lingshan Road, Pudong New Ddistrict, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.
| |
Collapse
|
35
|
Kyei-Barffour I, Margetts M, Vash-Margita A, Pelosi E. The Embryological Landscape of Mayer-Rokitansky-Kuster-Hauser Syndrome: Genetics and Environmental Factors. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2021; 94:657-672. [PMID: 34970104 PMCID: PMC8686787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a disorder caused by Müllerian ducts dysgenesis affecting 1 in 5000 women with a typical 46,XX karyotype. The etiology of MRKH syndrome is complex and largely unexplained. Familial clustering suggests a genetic component and the spectrum of clinical presentations seems consistent with an inheritance pattern characterized by incomplete penetrance and variable expressivity. Mutations of several candidate genes have been proposed as possible causes based on genetic analyses of human patients and animal models. In addition, studies of monozygotic twins with discordant phenotypes suggest a role for epigenetic changes following potential exposure to environmental compounds. The spectrum of clinical presentations is consistent with intricate disruptions of shared developmental pathways or signals during early organogenesis. However, the lack of functional validation and translational studies have limited our understanding of the molecular mechanisms involved in this condition. The clinical management of affected women, including early diagnosis, genetic testing of MRKH syndrome, and the implementation of counseling strategies, is significantly impeded by these knowledge gaps. Here, we illustrate the embryonic development of tissues and organs affected by MRKH syndrome, highlighting key pathways that could be involved in its pathogenesis. In addition, we will explore the genetics of this condition, as well as the potential role of environmental factors, and discuss their implications to clinical practice.
Collapse
Affiliation(s)
- Isaac Kyei-Barffour
- Department of Biomedical Sciences, University of Cape
Coast, Cape Coast, Ghana
| | - Miranda Margetts
- Center for American Indian and Rural Health Equity,
Montana State University, Bozeman, MT, USA
| | - Alla Vash-Margita
- Department of Obstetrics, Gynecology and Reproductive
Sciences, Division of Pediatric and Adolescent Gynecology, Yale University
School of Medicine, New Haven, CT, USA
| | - Emanuele Pelosi
- Centre for Clinical Research, The University of
Queensland, Brisbane, QLD, Australia
| |
Collapse
|
36
|
Li MH, Marty-Santos LM, van Ginkel PR, McDermott AE, Rasky AJ, Lukacs NW, Wellik DM. The Lung Elastin Matrix Undergoes Rapid Degradation Upon Adult Loss of Hox5 Function. Front Cell Dev Biol 2021; 9:767454. [PMID: 34901011 PMCID: PMC8662386 DOI: 10.3389/fcell.2021.767454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Hox genes encode transcription factors that are critical for embryonic skeletal patterning and organogenesis. The Hoxa5, Hoxb5, and Hoxc5 paralogs are expressed in the lung mesenchyme and function redundantly during embryonic lung development. Conditional loss-of-function of these genes during postnatal stages leads to severe defects in alveologenesis, specifically in the generation of the elastin network, and animals display bronchopulmonary dysplasia (BPD) or BPD-like phenotype. Here we show the surprising results that mesenchyme-specific loss of Hox5 function at adult stages leads to rapid disruption of the mature elastin matrix, alveolar enlargement, and an emphysema-like phenotype. As the elastin matrix of the lung is considered highly stable, adult disruption of the matrix was not predicted. Just 2 weeks after deletion, adult Hox5 mutant animals show significant increases in alveolar space and changes in pulmonary function, including reduced elastance and increased compliance. Examination of the extracellular matrix (ECM) of adult Tbx4rtTA; TetOCre; Hox5a f a f bbcc lungs demonstrates a disruption of the elastin network although the underlying fibronectin, interstitial collagen and basement membrane appear unaffected. An influx of macrophages and increased matrix metalloproteinase 12 (MMP12) are observed in the distal lung 3 days after Hox5 deletion. In culture, fibroblasts from Hox5 mutant lungs exhibit reduced adhesion. These findings establish a novel role for Hox5 transcription factors as critical regulators of lung fibroblasts at adult homeostasis.
Collapse
Affiliation(s)
- Mu-Hang Li
- Genetics Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Leilani M. Marty-Santos
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Paul R. van Ginkel
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Aubrey E. McDermott
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Andrew J. Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Deneen M. Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
37
|
Major AT, Estermann MA, Roly ZY, Smith CA. An evo-devo perspective of the female reproductive tract. Biol Reprod 2021; 106:9-23. [PMID: 34494091 DOI: 10.1093/biolre/ioab166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 01/22/2023] Open
Abstract
The vertebrate female reproductive tract has undergone considerable diversification over evolution, having become physiologically adapted to different reproductive strategies. This review considers the female reproductive tract from the perspective of evolutionary developmental biology (evo-devo). Very little is known about how the evolution of this organ system has been driven at the molecular level. In most vertebrates, the female reproductive tract develops from paired embryonic tubes, the Müllerian ducts. We propose that formation of the Müllerian duct is a conserved process that has involved co-option of genes and molecular pathways involved in tubulogenesis in the adjacent mesonephric kidney and Wolffian duct. Downstream of this conservation, genetic regulatory divergence has occurred, generating diversity in duct structure. Plasticity of the Hox gene code and wnt signaling, in particular, may underlie morphological variation of the uterus in mammals, and evolution of the vagina. This developmental plasticity in Hox and Wnt activity may also apply to other vertebrates, generating the morphological diversity of female reproductive tracts evident today.
Collapse
Affiliation(s)
- Andrew T Major
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Martin A Estermann
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Zahida Y Roly
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| |
Collapse
|
38
|
Ford MJ, Harwalkar K, Pacis AS, Maunsell H, Wang YC, Badescu D, Teng K, Yamanaka N, Bouchard M, Ragoussis J, Yamanaka Y. Oviduct epithelial cells constitute two developmentally distinct lineages that are spatially separated along the distal-proximal axis. Cell Rep 2021; 36:109677. [PMID: 34496237 DOI: 10.1016/j.celrep.2021.109677] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 05/25/2021] [Accepted: 08/17/2021] [Indexed: 01/02/2023] Open
Abstract
Owing to technical advances in single-cell biology, the appreciation of cellular heterogeneity has increased, which has aided our understanding of organ function, homeostasis, and disease progression. The oviduct (also known as the fallopian tube) is the distalmost portion of the female reproductive tract. It is essential for reproduction and the proposed origin of high-grade serous ovarian carcinoma (HGSOC). In mammals, the oviduct is morphologically segmented along the ovary-uterus axis into four evolutionally conserved regions. It is unclear, however, if there is a diversification of epithelial cell characteristics between these regions. In this study, we identify transcriptionally distinct populations of secretory and multiciliated cells restricted to the distal and proximal regions of the oviduct. We demonstrate that distal and proximal populations are distinct lineages specified early in Müllerian duct development and are maintained separately. These results aid our understanding of epithelial development, homeostasis, and initiation of disease from the oviduct.
Collapse
Affiliation(s)
- Matthew J Ford
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Keerthana Harwalkar
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Alain S Pacis
- Canadian Centre for Computational Genomics (C3G), Genome Quebec Innovation Centre, McGill University, Montreal, QC H3A 1A4, Canada
| | - Helen Maunsell
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Yu Chang Wang
- Department of Human Genetics, McGill University, Montreal, QC H3A OC7, Canada; McGill University and Genome Centre, Montreal, QC H3A 1A4, Canada
| | - Dunarel Badescu
- Department of Human Genetics, McGill University, Montreal, QC H3A OC7, Canada; McGill University and Genome Centre, Montreal, QC H3A 1A4, Canada
| | - Katie Teng
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Nobuko Yamanaka
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada
| | - Maxime Bouchard
- Rosalind and Morris Goodman Cancer Research Institute and Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University, Montreal, QC H3A OC7, Canada; McGill University and Genome Centre, Montreal, QC H3A 1A4, Canada; Department of Bioengineering, McGill University, Montreal, QC H3A 0C3, Canada
| | - Yojiro Yamanaka
- Rosalind and Morris Goodman Cancer Research Institute, Department of Human Genetics, McGill University, Montreal, QC H3A 1A3, Canada.
| |
Collapse
|
39
|
Zhu M, Yi S, Huang X, Meng J, Sun H, Zhou J. Human chorionic gonadotropin improves endometrial receptivity by increasing the expression of homeobox A10. Mol Hum Reprod 2021; 26:413-424. [PMID: 32502249 DOI: 10.1093/molehr/gaaa026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 03/20/2020] [Indexed: 12/25/2022] Open
Abstract
Homeobox A10 (HOXA10) is a characterized marker of endometrial receptivity. The mechanism by which hCG intrauterine infusion promotes embryo implantation is still unclear. This study seeks to investigate whether hCG improves endometrial receptivity by increasing expression of HOXA10. HOXA10 expression with human chorionic gonadotropin stimulation was analyzed in vitro and in vivo. Our results demonstrate that HOXA10 was decreased in the endometria of recurrent implantation failure patients compared to that in the healthy control fertile group, also we observed that hCG intrauterine infusion increased endometrial HOXA10 expression. HOXA10, blastocyst-like spheroid expansion area was increased, whereas DNA (cytosine-5-)-methyltransferase 1 was decreased when human endometrial stromal cells (hESCs) were treated with 0.2 IU/ml of hCG for 48 h. HOXA10 promoter methylation was also reduced after hCG treatment. Collagen XV (ColXV) can repress the expression of DNA (cytosine-5-)-methyltransferase 1, and hCG treatment increased the expression of ColXV. However, when the hESCs were treated with LH/hCG receptor small interfering RNA to knock down LH/hCG receptor, hCG treatment failed to repress DNA (cytosine-5-)-methyltransferase 1 expression or to increase ColXV expression. Our findings suggest that hCG may promote embryo implantation by increasing the expression of HOXA10.
Collapse
Affiliation(s)
- Mengchen Zhu
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Shanling Yi
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Xiaomin Huang
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Junan Meng
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Haixiang Sun
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Jianjun Zhou
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| |
Collapse
|
40
|
Albayrak M, Biyik I, Ozatik FY, Ozatik O, Ari NS, Teksen Y, Erten O. Cisplatin decreases HOXA13 and alphaVBeta3 integrin levels in the uterus. Taiwan J Obstet Gynecol 2021; 60:728-733. [PMID: 34247815 DOI: 10.1016/j.tjog.2021.05.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To examine the effects of cisplatin on uterine histology and implantation molecules and the possible protective role of recombinant Klotho administration on uterine histology and uterine receptivity in mice exposed to cisplatin. MATERIALS AND METHODS This study was conducted using thirty-two adult female mice assigned to four groups with 8 mice in each group. Saline was given to the 1st group, cisplatin to the 2nd group, recombinant mouse Klotho to the 3rd group and recombinant mouse Klotho plus cisplatin to the 4th group. Uterine tissues were examined for damage histologically and immunobiologically for the uterine receptivity markers HOXA13 and alphaVBeta3 integrin. RESULTS Apoptosis, degeneration, decrease in uterine thickness and uterine absence of gland scores were higher in the cisplatin group (3rd group) compared to the saline group (1st group) (cisplatin vs. saline p < 0.0001 for all parameters). In the recombinant Klotho plus cisplatin group (4th group), scores of apoptosis, degeneration, reduction in uterine thickness and uterine absence of gland were lower than the group receiving only cisplatin (cisplatin plus recombinant Klotho vs cisplatin, p = 0.006 for apoptosis; p = 0.017 for degeneration; p = 0.011 for the reduction in uterine thickness; p = 0.002 for the absence of gland). However, HOXA13 and alphaVBeta3 integrin staining levels were not different between the cisplatin group (group 3) and the cisplatin plus recombinant Klotho group (group 4) (p = 0.980 and p = 0.762, respectively.) CONCLUSION: Cisplatin has adverse effects on the uterus. Administration of recombinant Klotho was found to attenuate the cisplatin-induced damage but failed to preserve levels of the implantation molecules HOXA13 and alphaVbeta3. Further studies examining the effect of cisplatin toxicity using other implantation markers along with functional studies are needed.
Collapse
Affiliation(s)
- Mustafa Albayrak
- Florence Nightingale Hospital, Department of Obstetrics and Gynaecology, Istanbul, Turkey
| | - Ismail Biyik
- Kutahya Health Sciences University, School of Medicine, Department of Obstetrics and Gynaecology, Kutahya, Turkey.
| | - Fikriye Yasemin Ozatik
- Kutahya Health Sciences University, School of Medicine, Department of Medical Pharmacology, Kutahya, Turkey
| | - Orhan Ozatik
- Kutahya Health Sciences University, School of Medicine, Department of Histology and Embryology, Kutahya, Turkey
| | - Neziha Senem Ari
- Kutahya Health Sciences University, School of Medicine, Department of Histology and Embryology, Kutahya, Turkey
| | - Yasemin Teksen
- Kutahya Health Sciences University, School of Medicine, Department of Medical Pharmacology, Kutahya, Turkey
| | - Ozlem Erten
- Florence Nightingale Hospital, Department of Obstetrics and Gynaecology, Istanbul, Turkey
| |
Collapse
|
41
|
Santana González L, Artibani M, Ahmed AA. Studying Müllerian duct anomalies - from cataloguing phenotypes to discovering causation. Dis Model Mech 2021; 14:269240. [PMID: 34160006 PMCID: PMC8246269 DOI: 10.1242/dmm.047977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Müllerian duct anomalies (MDAs) are developmental disorders of the Müllerian duct, the embryonic anlage of most of the female reproductive tract. The prevalence of MDAs is 6.7% in the general female population and 16.7% in women who exhibit recurrent miscarriages. Individuals affected by these anomalies suffer from high rates of infertility, first-trimester pregnancy losses, premature labour, placental retention, foetal growth retardation and foetal malpresentations. The aetiology of MDAs is complex and heterogeneous, displaying a range of clinical pictures that generally lack a direct genotype-phenotype correlation. De novo and familial cases sharing the same genomic lesions have been reported. The familial cases follow an autosomal-dominant inheritance, with reduced penetrance and variable expressivity. Furthermore, few genetic factors and molecular pathways underpinning Müllerian development and dysregulations causing MDAs have been identified. The current knowledge in this field predominantly derives from loss-of-function experiments in mouse and chicken models, as well as from human genetic association studies using traditional approaches, such as microarrays and Sanger sequencing, limiting the discovery of causal factors to few genetic entities from the coding genome. In this Review, we summarise the current state of the field, discuss limitations in the number of studies and patient samples that have stalled progress, and review how the development of new technologies provides a unique opportunity to overcome these limitations. Furthermore, we discuss how these new technologies can improve functional validation of potential causative alterations in MDAs. Summary: Here, we review the current knowledge about Müllerian duct anomalies in the context of new high-throughput technologies and model systems and their implications in the prevention of these disorders.
Collapse
Affiliation(s)
- Laura Santana González
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Mara Artibani
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Ahmed Ashour Ahmed
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
42
|
Nuclear expression of VDR and AHR is mutually exclusive in glandular cells in endometriosis. Histochem Cell Biol 2021; 156:391-399. [PMID: 34155552 PMCID: PMC8550147 DOI: 10.1007/s00418-021-02005-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 11/03/2022]
Abstract
The vitamin D receptor (VDR) and aryl hydrocarbon receptor (AHR) are two nuclear receptors that exert their effects by binding with ligands and forming a molecular complex. These complexes translocate to the nucleus and activate the expression of a series of genes which have a response element to VDR or AHR. Both receptors have been identified in the pathogenesis of endometriosis, a common disease characterized by the formation of endometrium-like tissue in ectopic zones. Despite numerous therapies, there is no definitive cure for endometriosis at the pharmacological level. Our study aims to describe the location and the expression of VDR and AHR at the protein level. For this purpose, an evaluation was performed using tissue from the three normal phases of the endometrium (proliferative, early, and late secretory) and in endometriosis by immunohistochemistry, using anti-VDR and anti-AHR antibodies. We demonstrate that in the nuclei of glandular cells in endometriosis, the expression of VDR and AHR is mutually exclusive—when the expression of one receptor is high, the other one is low—suggesting a possible target in the treatment of endometriosis. We also identify a significant change in the expression of glandular cytoplasmic AHR between the proliferative and late secretory endometrium.
Collapse
|
43
|
Xu B, Geerts D, Hu S, Yue J, Li Z, Zhu G, Jin L. The depot GnRH agonist protocol improves the live birth rate per fresh embryo transfer cycle, but not the cumulative live birth rate in normal responders: a randomized controlled trial and molecular mechanism study. Hum Reprod 2021; 35:1306-1318. [PMID: 32478400 DOI: 10.1093/humrep/deaa086] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Do cumulative live birth rates (CLBRs) after one complete ART cycle differ between the three commonly used controlled ovarian stimulation (COS) protocols (GnRH antagonist, depot GnRHa (GnRH agonist) and long GnRHa) in normal responders undergoing IVF/ICSI? SUMMARY ANSWER There were similar CLBRs between the GnRH antagonist, depot GnRHa and long GnRHa protocols. WHAT IS KNOWN ALREADY There is no consensus on which COS protocol is the most optimal in women with normal ovarian response. The CLBR provides the final success rate after one complete ART cycle, including the fresh and all subsequent frozen-thawed embryo transfer (ET) cycles. We suggest that the CLBR measure would allow for better comparisons between the different treatment protocols. STUDY DESIGN, SIZE, DURATION A prospective controlled, randomized, open label trial was performed between May 2016 and May 2017. A total of 819 patients were allocated to the GnRH antagonist, depot GnRHa or long GnRHa protocol in a 1:1:1 ratio. The minimum follow-up time from the first IVF cycle was 2 years. To further investigate the potential effect of COS with the GnRH antagonist, depot GnRHa or long GnRHa protocol on endometrial receptivity, the expression of homeobox A10 (HOXA10), myeloid ecotropic viral integration site 1 (MEIS1) and leukemia inhibitory factor (LIF) endometrial receptivity markers was evaluated in endometrial tissue from patients treated with the different COS protocols. PARTICIPANTS/MATERIALS, SETTING, METHODS Infertile women with normal ovarian response (n = 819) undergoing IVF/ICSI treatment were randomized to the GnRH antagonist, depot GnRHa or long GnRHa protocol. Both IVF and ICSI cycles were included, and the sperm samples used were either fresh or frozen partner ejaculates or frozen donor ejaculates. The primary outcome was the live birth rate (LBR) per fresh ET cycle, and the CLBR after one complete ART cycle, until the birth of a first child (after 28 weeks) or until all frozen embryos were used, whichever occurred first. Pipelle endometrial biopsies from 34 female patients were obtained on Days 7-8 after oocyte retrieval or spontaneous ovulation in natural cycles, respectively, and HOXA10, MEIS1 and LIF mRNA and protein expression levels in the human endometrium was determined by quantitative real-time PCR and western blot, respectively. MAIN RESULTS AND THE ROLE OF CHANCE There were no significant differences in CLBRs between the GnRH antagonist, depot GnRHa or long GnRHa protocol (71.4 versus 75.5 versus 72.2%, respectively). However, there was a significantly higher LBR per fresh ET cycle in the depot GnRHa protocol than in the long GnRHa and GnRH antagonist protocols (62.6 versus 52.1% versus 45.6%, P < 0.05). Furthermore, HOXA10, MEIS1 and LIF mRNA and protein expression in endometrium all showed significantly higher in the depot GnRHa protocol than in the long GnRHa and GnRH antagonist protocols (P < 0.05). LIMITATIONS, REASONS FOR CAUTION A limitation of our study was that both our clinicians and patients were not blinded to the randomization for the randomized controlled trial (RCT). An inclusion criterion for the current retrospective cohort study was based on the 'actual ovarian response' during COS treatment, while the included population for the RCT was 'expected normal responders' based on maternal age and ovarian reserve test. In addition, the analysis was restricted to patients under 40 years of age undergoing their first IVF cycle. Furthermore, the endometrial tissue was collected from patients who cancelled the fresh ET, which may include some patients at risk for ovarian hyperstimulation syndrome, however only patients with 4-19 oocytes retrieved were included in the molecular study. WIDER IMPLICATIONS OF THE FINDINGS The depot GnRH agonist protocol improves the live birth rate per fresh ET cycle, but not the cumulative live birth rate in normal responders. A possible explanation for the improved LBR after fresh ET in the depot GnRHa protocol could be molecular signalling at the level of endometrial receptivity. STUDY FUNDING/COMPETING INTEREST(S) This project was funded by Grant 81571439 from the National Natural Sciences Foundation of China and Grant 2016YFC1000206-5 from the National Key Research & Development Program of China. The authors declare no conflict of interest. TRIAL REGISTRATION NUMBER The RCT trial was registered at the Chinese Clinical Trial Registry, Study Number: ChiCTR-INR-16008220. TRIAL REGISTRATION DATE 5 April 2016. DATE OF FIRST PATIENT’S ENROLLMENT 12 May 2016.
Collapse
Affiliation(s)
- Bei Xu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan 430030, People's Republic of China
| | - Dirk Geerts
- Department of Medical Biology, Amsterdam University Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Shiqiao Hu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan 430030, People's Republic of China
| | - Jing Yue
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan 430030, People's Republic of China
| | - Zhou Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan 430030, People's Republic of China
| | - Guijin Zhu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan 430030, People's Republic of China
| | - Lei Jin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan 430030, People's Republic of China
| |
Collapse
|
44
|
Santana Gonzalez L, Rota IA, Artibani M, Morotti M, Hu Z, Wietek N, Alsaadi A, Albukhari A, Sauka-Spengler T, Ahmed AA. Mechanistic Drivers of Müllerian Duct Development and Differentiation Into the Oviduct. Front Cell Dev Biol 2021; 9:605301. [PMID: 33763415 PMCID: PMC7982813 DOI: 10.3389/fcell.2021.605301] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
The conduits of life; the animal oviducts and human fallopian tubes are of paramount importance for reproduction in amniotes. They connect the ovary with the uterus and are essential for fertility. They provide the appropriate environment for gamete maintenance, fertilization and preimplantation embryonic development. However, serious pathologies, such as ectopic pregnancy, malignancy and severe infections, occur in the oviducts. They can have drastic effects on fertility, and some are life-threatening. Despite the crucial importance of the oviducts in life, relatively little is known about the molecular drivers underpinning the embryonic development of their precursor structures, the Müllerian ducts, and their successive differentiation and maturation. The Müllerian ducts are simple rudimentary tubes comprised of an epithelial lumen surrounded by a mesenchymal layer. They differentiate into most of the adult female reproductive tract (FRT). The earliest sign of Müllerian duct formation is the thickening of the anterior mesonephric coelomic epithelium to form a placode of two distinct progenitor cells. It is proposed that one subset of progenitor cells undergoes partial epithelial-mesenchymal transition (pEMT), differentiating into immature Müllerian luminal cells, and another subset undergoes complete EMT to become Müllerian mesenchymal cells. These cells invaginate and proliferate forming the Müllerian ducts. Subsequently, pEMT would be reversed to generate differentiated epithelial cells lining the fully formed Müllerian lumen. The anterior Müllerian epithelial cells further specialize into the oviduct epithelial subtypes. This review highlights the key established molecular and genetic determinants of the processes involved in Müllerian duct development and the differentiation of its upper segment into oviducts. Furthermore, an extensive genome-wide survey of mouse knockout lines displaying Müllerian or oviduct phenotypes was undertaken. In addition to widely established genetic determinants of Müllerian duct development, our search has identified surprising associations between loss-of-function of several genes and high-penetrance abnormalities in the Müllerian duct and/or oviducts. Remarkably, these associations have not been investigated in any detail. Finally, we discuss future directions for research on Müllerian duct development and oviducts.
Collapse
Affiliation(s)
- Laura Santana Gonzalez
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ioanna A Rota
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Developmental Immunology Research Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Mara Artibani
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matteo Morotti
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Zhiyuan Hu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Nina Wietek
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Abdulkhaliq Alsaadi
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ashwag Albukhari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tatjana Sauka-Spengler
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ahmed A Ahmed
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Liu L, Cheng J, Wei F, Pang L, Zhi Z, Yang W, Tan W. Lnc-RNA LINC01279 induces endometriosis via targeting of HOXA10. J Obstet Gynaecol Res 2021; 47:1825-1836. [PMID: 33657670 DOI: 10.1111/jog.14723] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 12/28/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
AIM To explore the regulatory role and molecular mechanism of lncRNA-LINC01279 in endometriosis (EMs). METHODS Between September 2018 and July 2019, 20 EMs patients and 20 healthy subjects were recruited to detect the expression of lncRNA-LINC01279 in EMs and in normal endometrium via qRT-PCR. Autograft was used to establish EMs models on Spraque-Dawley (SD) rats, which was followed by taking volume measurements of EMs endometrium and observing pathological changes in the morphology of EMs via hematoxylin and eosin (H&E) staining. The qRT-PCR technique was further carried out to determine mRNA expression of lncRNA-LINC01279 and HOXA10 in the serum of EMs rats and LINC01279 shRNA-transfected rats, while the protein expression of HOXA10 was determined using a Western blot. RESULTS EMs patients presented with upregulation of lncRNA-LINC01279 and downregulation of HOXA10 (p < 0.01 or 0.001). Online predictions further revealed that lncRNA-LINC01279 regulated the expression of HOXA10 via miRNA-135b. In EMs models, it was observed that there were a significantly enlarged endometrium and poor pathological morphology, significant upregulation of lncRNA-LINC01279, and downregulation of miR-135b and HOXA10 in serum (p < 0.05, 0.01 or 0.001). In the lncRNA-LINC01279 shRNA group, EMs rats, following treatment, had a sharp decrease in the volume of EMs endometrium, and an improvement in pathological morphology, while lncRNA-LINC01279 was downregulated, with upregulation of miR-135b and HOXA10 (p < 0.05, 0.01 or 0.001). CONCLUSION LncRNA-LINC01279, by the mechanism of targeting miR-135b, has the potential to downregulate the expression of HOXA10, and therefore, can promote the development and progression of EMs.
Collapse
Affiliation(s)
- Liling Liu
- Department of Reproductive Medicine and Genetics Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Junping Cheng
- Department of Reproductive Medicine and Genetics Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Fu Wei
- Department of Reproductive Medicine and Genetics Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Lihong Pang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of GuangXi Medical University, Nanning, Guangxi, China
| | - Zhifu Zhi
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of GuangXi Medical University, Nanning, Guangxi, China
| | - Wenmei Yang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of GuangXi Medical University, Nanning, Guangxi, China
| | - Weihong Tan
- Department of Reproductive Medicine and Genetics Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| |
Collapse
|
46
|
Alderman MH, Taylor HS. Molecular mechanisms of estrogen action in female genital tract development. Differentiation 2021; 118:34-40. [PMID: 33707128 DOI: 10.1016/j.diff.2021.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 10/22/2022]
Abstract
The development of the female reproductive tract can be divided into three parts consisting of Müllerian duct organogenesis, pre-sexual maturation organ development, and post-sexual maturation hormonal regulation. In primates, Müllerian duct organogenesis proceeds in an estrogen independent fashion based on transcriptional pathways that are suppressed in males by the presence of AMH and SRY. However, clinical experience indicates that exposure to xenoestrogens such as diethylstilbestrol (DES) during critical periods including late organogenesis and pre-sexual maturational development can have substantial effects on uterine morphology, and confer increased risk of disease states later in life. Recent evidence has demonstrated that these effects are in part due to epigenetic regulation of gene expression, both in the form of aberrant CpG methylation, and accompanying histone modifications. While xenoestrogens and selective estrogen receptor modulators (SERMS) both can induce non-canonical binding confirmations in estrogen receptors, the primate specific fetal estrogens Estriol and Estetrol may act in a similar fashion to alter gene expression through tissue specific epigenetic modulation.
Collapse
Affiliation(s)
- Myles H Alderman
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, 06520, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
47
|
Effects of Erbuzhuyu Decoction Combined with Acupuncture on Endometrial Receptivity Are Associated with the Expression of miR-494-3p. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9739672. [PMID: 33299465 PMCID: PMC7710412 DOI: 10.1155/2020/9739672] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/23/2020] [Accepted: 11/09/2020] [Indexed: 11/17/2022]
Abstract
Background/Aim Erbuzhuyu decoction (EBZYD) is a traditional Chinese medicine (TCM) formula and has been used in infertility treatment. Meanwhile, acupuncture is also used to treat female infertility. However, it is unclear whether EBZYD combined with acupuncture has better therapeutic effect. The aim of this study was to explore the effect of EBZYD combined with acupuncture and investigate its mechanism in superovulation mice. Methods The mice received the treatment of EBZYD, acupuncture, EBZYD combined with acupuncture, or miR-494-3p agomir combined with EBZYD and acupuncture. The blastocysts' number, endometrial microstructure, and endometrial thickness were observed, followed by the detection of endometrial receptivity-related factors, PI3K/Akt/mTOR pathway-related proteins, and miR-494-3p expression using quantitative real-time polymerase chain reaction (qRT-PCR) or western blot. Luciferase reporter assay was performed to confirm the targeting relationship between HOXA10 and miR-494-3p. Results EBZYD combined with acupuncture treatment could increase the number of blastocysts, pinopodes, endometrial thickness, and the expression of endometrial receptivity-related factors, and the treatment effect of EBZYD combined with acupuncture was better than EBZYD or acupuncture alone. In addition, EBZYD combined with acupuncture treatment activated PI3K/Akt/mTOR pathway and inhibited the expression of miR-494-3p. HOXA10 is one of the target genes of miR-494-3p. Overexpression of miR-494-3p reversed the therapeutic effect of EBZYD combined with acupuncture and suppressed the expression of HOXA10 and the activity of PI3K/Akt/mTOR pathway. Conclusion This study suggests that EBZYD combined with acupuncture could improve endometrial receptivity in superovulation mice via miR-494-3p/HOXA10 axis.
Collapse
|
48
|
Menichini D, Forte G, Orrù B, Gullo G, Unfer V, Facchinetti F. The role of vitamin D in metabolic and reproductive disturbances of polycystic ovary syndrome: A narrative mini-review. INT J VITAM NUTR RES 2020; 92:126-133. [PMID: 33284035 DOI: 10.1024/0300-9831/a000691] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Vitamin D is a secosteroid hormone that plays a pivotal role in several metabolic and reproductive pathways in humans. Increasing evidence supports the role of vitamin D deficiency in metabolic disturbances and infertility in women with polycystic ovary syndrome (PCOS). Indeed, supplementation with vitamin D seems to have a beneficial role on insulin resistance and endometrial receptivity. On the other hand, exceedingly high levels of vitamin D appear to play a detrimental role on oocytes development and embryo quality. In the current review, we summarize the available evidence about the topic, aiming to suggest the best supplementation strategy in women with PCOS or, more generally, in those with metabolic disturbances and infertility. Based on the retrieved data, vitamin D seems to have a beneficial role on IR, insulin sensitivity and endometrial receptivity, but high levels and incorrect timing of administration seem to have a detrimental role on oocytes development and embryo quality. Therefore, we encourage a low dose supplementation (400-800 IU/day) particularly in vitamin D deficient women that present metabolic disturbances like PCOS. As far as the reproductive health, we advise vitamin D supplementation in selected populations, only during specific moments of the ovarian cycle, to support the luteal phase. However, ambiguities about dosage and timing of the supplementation still emerge from the clinical studies published to date and further studies are required.
Collapse
Affiliation(s)
- Daniela Menichini
- International Doctorate School in Clinical and Experimental Medicine, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy.,Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, Italy
| | | | - Beatrice Orrù
- Medical Affairs Department, Lo.Li. Pharma, Rome, Italy
| | - Giuseppe Gullo
- IVF Public Center, AOOR Villa Sofia Cervello, University Hospital, Palermo, Italy
| | - Vittorio Unfer
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Fabio Facchinetti
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, Italy
| |
Collapse
|
49
|
Nameki R, Chang H, Reddy J, Corona RI, Lawrenson K. Transcription factors in epithelial ovarian cancer: histotype-specific drivers and novel therapeutic targets. Pharmacol Ther 2020; 220:107722. [PMID: 33137377 DOI: 10.1016/j.pharmthera.2020.107722] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
Transcription factors (TFs) are major contributors to cancer risk and somatic development. In preclinical and clinical studies, direct or indirect inhibition of TF-mediated oncogenic gene expression profiles have proven to be effective in many tumor types, highlighting this group of proteins as valuable therapeutic targets. In spite of this, our understanding of TFs in epithelial ovarian cancer (EOC) is relatively limited. EOC is a heterogeneous disease composed of five major histologic subtypes; high-grade serous, low-grade serous, endometrioid, clear cell and mucinous. Each histology is associated with unique clinical etiologies, sensitivity to therapies, and molecular signatures - including diverse transcriptional regulatory programs. While some TFs are shared across EOC subtypes, a set of TFs are expressed in a histotype-specific manner and likely explain part of the histologic diversity of EOC subtypes. Targeting TFs present with unique opportunities for development of novel precision medicine strategies for ovarian cancer. This article reviews the critical TFs in EOC subtypes and highlights the potential of exploiting TFs as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Robbin Nameki
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Heidi Chang
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jessica Reddy
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rosario I Corona
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kate Lawrenson
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Center for Bioinformatics and Functional Genomics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
50
|
Zhang L, Dai F, Chen G, Wang Y, Liu S, Zhang L, Xian S, Yuan M, Yang D, Zheng Y, Deng Z, Cheng Y, Yang X. Molecular mechanism of extracellular matrix disorder in pelvic organ prolapses. Mol Med Rep 2020; 22:4611-4618. [PMID: 33173982 PMCID: PMC7646844 DOI: 10.3892/mmr.2020.11564] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022] Open
Abstract
Pelvic organ prolapses (POP) notably reduces the quality of life in elderly populations due to bladder and bowel dysfunction, incontinence, and coital problems. Extracellular matrix (ECM) disorder is a pivotal event in the progression of POP, but to date, its specific underlying mechanism remains unclear. The ligaments of patients with POP and healthy controls were collected to compare the expression of Homeobox11 (HOXA11) and transforming growth factor β (TGF-β1) via immunohistochemical analysis. HOXA11 and TGF-β1 were overexpressed or knocked down in fibroblast cells to explore their effects on the expression of collagen and matrix metalloproteinases (MMPs). HOXA11 and TGF-β1 were greatly reduced in the ligaments of patients with POP. The overexpression and downregulation of HOXA11 and TGF-β1 can mediate ECM disorder via regulating expression of collagen (Col) and MMPs. In addition, HOXA11 and TGF-β1 exerted synergistic effect on the expression of Col and MMPs. The present study identified that HOXA11 and TGF-β1 serve critical roles in mediating ECM disorders, which may be of clinical significance for the diagnosis and treatment of patients with POP.
Collapse
Affiliation(s)
- Liping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Gantao Chen
- Department of Gastroenterology, Third People's Hospital of Xiantao in Hubei Province, Xiantao, Hubei 433000, P.R. China
| | - Yanqing Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Li Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shu Xian
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mengqin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yajing Zheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaofeng Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|