1
|
da Silva TQM, Barbosa EM, Santos LC, de Oliveira LS, Cunha MCDSG, de Macedo IO, Martins BGC, Oliveira CL, Rodrigues NP, Araújo-Lopes R, Szawka RE, Silva JF. Hypothyroidism Alters Uterine Kisspeptin System and Activity Modulators in Cyclic Rats. Int J Mol Sci 2025; 26:543. [PMID: 39859259 PMCID: PMC11765193 DOI: 10.3390/ijms26020543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Hypothyroidism causes ovarian dysfunction and infertility in women and animals and impairs the hypothalamic expression of kisspeptin (Kp). However, kisspeptin is also expressed in the genital system, and the lack of the Kp receptor (Kiss1r) in the uterus is linked to reduced implantation rates. This study investigated the impact of hypothyroidism on the uterine expression of Kp and Kiss1r in female rats throughout the estrous cycle and the associated changes in uterine activity modulators. Hypothyroidism was induced through daily administration of propylthiouracil (PTU) over a period of 14 days. Plasma levels of LH, E2, and P4, cyclicity, body and uterine weight, uterine histomorphometry, and the gene and/or protein expression of Kiss1, Kiss1r, estrogen receptor α (ERα), progesterone receptor (PR), and thyroid hormone receptor α (TRα) were assessed. Additionally, proliferative activity (CDC-47) and the gene expression of uterine receptivity mediators (SMO, WNT4, BMP2, HAND2, MUC1, and LIF) were evaluated. Hypothyroidism prolonged the diestrus and increased progesterone levels during this phase, while decreasing luteinizing hormone and estradiol on proestrus. In the uterus, hypothyroidism reduced Kp immunostaining on diestrus and KISS1R mRNA levels on proestrus. These changes were accompanied by reduced endometrial glands, reduced uterine proliferative activity, and reduced ERα gene and protein expression. Additionally, hypothyroidism led to reduced uterine gene expression of LIF, BMP2, WNT4, and HAND2. On the other hand, thyroid hypofunction increased uterine PR and TRα immunostaining, while it reduced PGR gene expression on diestrus. These findings demonstrate that hypothyroidism reduces the expression of Kiss1/Kiss1r system in the uterus, which is associated with disrupted uterine estrogen and progesterone signaling and reduced expression of uterine receptivity mediators across the rat estrous cycle.
Collapse
Affiliation(s)
- Thayná Queiroz Menezes da Silva
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Erikles Macêdo Barbosa
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Luciano Cardoso Santos
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Luciana Santos de Oliveira
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Maria Clara da Silva Galrão Cunha
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Isabella Oliveira de Macedo
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Brenda Geovana Campos Martins
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Cibele Luz Oliveira
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Natalia Panhoca Rodrigues
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| | - Roberta Araújo-Lopes
- Endocrinology and Metabolism Laboratory, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (R.A.-L.); (R.E.S.)
| | - Raphael Escorsim Szawka
- Endocrinology and Metabolism Laboratory, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (R.A.-L.); (R.E.S.)
| | - Juneo Freitas Silva
- Electron Microscopy Center, Department of Biological Sciences, State University of Santa Cruz, Ilheus 45662-900, Brazil; (T.Q.M.d.S.); (E.M.B.); (L.C.S.); (L.S.d.O.); (M.C.d.S.G.C.); (I.O.d.M.); (B.G.C.M.); (C.L.O.); (N.P.R.)
| |
Collapse
|
2
|
Bedir Ö, Tavares Pereira M, Rehrauer H, Grazul-Bilska A, Kowalewski MP. Transcriptomic alterations in the ovine caruncular endometrium due to imbalanced nutrition and FSH-induced ovarian hyperstimulation. BMC Genomics 2024; 25:1216. [PMID: 39695382 DOI: 10.1186/s12864-024-10799-w] [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: 04/15/2024] [Accepted: 09/12/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Imbalanced diet and exogenous gonadotrophins affect uterine function and morphology. In sheep, FSH-induced superovulation alters implantation-related gene expression, influenced by both treatment and diet. In this study, we used deep RNA sequencing (NGS, RNA-Seq) to expand our understanding of these effects on the caruncular endometrium. METHODS Ewes (n = 3-5/group) were separated into control fed (CF), overfed (OF), and underfed (UF) groups, with each group subdivided between FSH (superovulated; SOV) or saline (negative controls; CONT) treatment. Caruncular samples were collected on day 10 of diestrus of the subsequent estrous cycle, with samples from CF_CONT also collected on day 5 to assess time-dependent changes. RESULTS The 1484 differentially expressed genes (DEGs, P < 0.01, FDR < 0.05) identified between CF_CONT animals at days 5 and 10 were predominantly associated with increased immune activity and cellular metabolic processes and cellular proliferation. In CONT animals, imbalanced nutrition (i.e., both OF and UF) was associated with enrichment of terms associated with cell adhesion and differentiation, immune response and angiogenesis. The FSH carry-over effects resulted in a higher number of DEGs in CF animals (1374), than in OF (168) or UF (18), mostly associated with dysregulation of cell cycle and hormonal sensitivity. CONCLUSION The absence of genes concurrently affected by superovulation (SOV) in all feeding regimes indicates that the effects of FSH on the caruncular transcriptome are multidirectional and dependent upon body condition. Therefore, the homeostasis of ovine caruncles is influenced by both body condition and superovulation (SOV), potentially affecting uterine receptivity.
Collapse
Affiliation(s)
- Özlem Bedir
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Winterthurerstrasse 260, Zürich, CH-8057, Switzerland
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Harran University, Sanliurfa, Turkey
| | - Miguel Tavares Pereira
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Winterthurerstrasse 260, Zürich, CH-8057, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich (FGCZ), ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Anna Grazul-Bilska
- Department of Animal Sciences, North Dakota State University, Fargo, USA
| | - Mariusz P Kowalewski
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Winterthurerstrasse 260, Zürich, CH-8057, Switzerland.
- Center for Clinical Studies (ZKS), Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland.
| |
Collapse
|
3
|
Wang Y, Li Q, He P, Zhang L, Chao T, Wang J. The TCONS_00046732/chi-miR-135b-5p/PRLR regulatory axis promotes endometrial epithelial cells growth through the PI3K-Akt signaling pathway. Int J Biol Macromol 2024; 283:137248. [PMID: 39505183 DOI: 10.1016/j.ijbiomac.2024.137248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Non-coding RNAs are considered key regulatory factors in biological and reproductive physiological processes in mammals. However, the molecular functions of long noncoding RNAs (lncRNAs) in regulating dynamic uterine development and reproductive capacity during sexual maturation remain unclear. This study analyzed the expression characteristics and molecular functions of lncRNAs in uterine tissues from 20 goats at four specific time points during sexual maturation: day 1 after birth (D1), 2 months (M2), 4 months (M4), and 6 months (M6), finding that stage-specific DE lncRNAs may regulate cell proliferation, apoptosis, hormone secretion, metabolism, and immune response through multiple action modes. Within the lncRNA-miRNA-mRNA network, a novel lncRNA, TCONS_00046732, associated with uterine development, exhibited significantly higher expression during sexual maturity compared to the prepubertal stage, correlating positively with PRLR and negatively with chi-miR-135b-5p. FISH and IF analyses revealed significant co-localization and distinct expression patterns of TCONS_00046732, chi-miR-135b-5p, and PRLR in the endometrial epithelium. Further experiments confirmed that TCONS_00046732 competitively binds to chi-miR-135b-5p to upregulate PRLR, thereby activating the PI3K-Akt signaling pathway, promoting primary endometrial epithelial cell proliferation, G1-to-S phase transition, and inhibiting apoptosis. These findings enhance our understanding of uterine molecular regulation and provide key insights into the molecular basis of goat sexual development.
Collapse
Affiliation(s)
- Yanyan Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Qing Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Peipei He
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China.
| | - Lu Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Tianle Chao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China.
| | - Jianmin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China.
| |
Collapse
|
4
|
Zhang X, Zhang L, Li T, Zhang Z, Shang X, Bai H, Liu Y, Zong X, Shang C, Song D, Zhang X, Fan L, Liu Z. Investigating bacteria-induced inflammatory responses using novel endometrial epithelial gland organoid models. FRONTIERS IN REPRODUCTIVE HEALTH 2024; 6:1490520. [PMID: 39600797 PMCID: PMC11588683 DOI: 10.3389/frph.2024.1490520] [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: 09/12/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction The endometrium plays a crucial role in early human pregnancy, particularly in embryo implantation, survival, and growth. However, invasion and infection by pathogens can lead to endometritis, infertility, and poor reproductive outcomes. Understanding the mechanisms of endometritis and its impact on fertility remains limited. An infection model using patient-derived endometrial epithelial gland organoids (EEGOs) was established to advance in vitro studies on endometritis and related infertility. Methods An EEGOs infection model was constructed and characterized from human endometrium, treating the organoids with estrogen and progesterone to observe changes in the proliferative and secretory phases. The organoids were infected with E. coli, and the release of inflammatory cytokines in the supernatant was detected using ELISA. RNA-seq was employed to analyze the differences before and after E. coli treatment, and differential gene mRNA expression was validated using real-time quantitative PCR. Additionally, the effect of E2 in alleviating inflammation was assessed through markers of receptivity (PAEP, LIF, ITGβ), proliferation (Ki67), and barrier repair (ZO-1). Results The constructed human EEGOs exhibited long-term expansion capability, genetic stability, and characteristic hormonal responses, strongly expressing epithelial markers (MUC1, E-Cadherin). After E. coli infection, the expression levels of inflammatory cytokines TNF-α, IL-8, and IFN-γ increased significantly (P < 0.05). RNA-seq indicated that the MAPK signaling pathway was activated post-infection, with increased expression levels of heat shock proteins and transcription factor mRNA. E2 treatment post-infection significantly decreased the mRNA expression of inflammatory genes IL-1β, IL8, IL6 and TNF-α compared to the E. coli infected group (P < 0.05). Additionally, the expression of genes related to receptivity, proliferation, and barrier repair was enhanced in the E2-treated organoids. Conclusions Our findings demonstrate that patient-derived EEGOs are responsive to bacterial infection and are effective models for studying host-pathogen interactions in bacterial infections. These organoids revealed the anti-inflammatory potential of E2 in alleviating E. coli-induced inflammation, providing insights into the mechanisms of endometritis and its impact on infertility. The study supports the use of EEGOs as valuable tools for understanding endometrial health and developing targeted treatments.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Li Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Ting Li
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Zhan Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xiang Shang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Huihui Bai
- Department of Clinical Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yong Liu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xiaonan Zong
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Chenguang Shang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Dan Song
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xu Zhang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Linyuan Fan
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Zhaohui Liu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| |
Collapse
|
5
|
Cassiani AG, Aloia TPA, Sousa-Vidal ÉK, Podgaec S, Piccinato CDA, Serrano-Nascimento C. Prenatal exposure to nitrate alters uterine morphology and gene expression in adult female F1 generation rats. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240085. [PMID: 39876961 PMCID: PMC11771761 DOI: 10.20945/2359-4292-2024-0085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/12/2024] [Indexed: 01/31/2025]
Abstract
Objective Nitrate is ubiquitously found in the environment and is one of the main components of nitrogen fertilizers. Previous studies have shown that nitrate disrupts the reproductive system in aquatic animals, but no study has evaluated the impact of nitrate exposure on the uterus in mammals. This study aimed to evaluate the impact of maternal exposure to nitrate during the prenatal period on uterine morphology and gene expression in adult female F1 rats. Materials and methods Pregnant Wistar rats were either treated with sodium nitrate 20 mg/L or 50 mg/L dissolved in drinking water from the first day of pregnancy until the birth of the offspring or were left untreated. On postnatal day 90, the uteri of female offspring rats were collected for histological and gene expression analyses. Morphometric analyses of the uterine photomicrographs were performed to determine the thickness of the layers of the uterine wall (endometrium, myometrium, and perimetrium) and the number of endometrial glands. Results The highest nitrate dose increased the myometrial thickness of the exposed female rats. Treatment with both nitrate doses reduced the number of endometrial glands compared with no treatment. Additionally, nitrate treatment significantly increased the expression of estrogen receptors and reduced the expression of progesterone receptors in the uterus. Conclusion Our results strongly suggest that prenatal exposure to nitrate programs gene expression and alters the uterine morphology in female F1 rats, potentially increasing their susceptibility to developing uterine diseases during adulthood.
Collapse
Affiliation(s)
- André Gilberto Cassiani
- Hospital Israelita Albert EinsteinSão PauloSPBrasilHospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | | | - Érica Kássia Sousa-Vidal
- Hospital Israelita Albert EinsteinSão PauloSPBrasilHospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | - Sérgio Podgaec
- Hospital Israelita Albert EinsteinSão PauloSPBrasilHospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | - Carla de Azevedo Piccinato
- Hospital Israelita Albert EinsteinSão PauloSPBrasilHospital Israelita Albert Einstein, São Paulo, SP, Brasil
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoDepartamento de Ginecologia e ObstetríciaRibeirão PretoSPBrasilDepartamento de Ginecologia e Obstetrícia da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | | |
Collapse
|
6
|
Yang M, Wang K, Zhang L, Zhang H, Zhang C. DCAF2 is essential for the development of uterine epithelia and mouse fertility. Front Cell Dev Biol 2024; 12:1474660. [PMID: 39364135 PMCID: PMC11446810 DOI: 10.3389/fcell.2024.1474660] [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: 08/02/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Introduction The successful outcome of a pregnancy depends on the proper functioning uterine epithelium. DNA damage binding protein 1 and cullin 4-associated factor 2 (DCAF2), a conserved substrate receptor for the cullin 4-RING E3 ubiquitin ligase (CRL4) complex, is essential for maintaining genome stability by facilitating ubiquitin-mediated degradation of substrates. Methods To better understand the physiological role of DCAF2 in female reproduction, we conducted a study using mice with conditional knockout (cKO) of DCAF2 in the uterus using the progesterone receptor Cre (Pgr Cre/+) mouse model. Results Our results showed the cKO mice were completely infertile, despite having ovarian function. The cKO mice exhibited severely thin uteri, demonstrating notable defects in both the uterine epithelium and a lack of glands. In addition, there were impaired proliferation and differentiation of epithelial cells in the cKO mice, ultimately resulting in failed implantation. Moreover, through deciphering the uterine transcriptome of cKO mice, we revealed crucial differentially expressed genes associated with steroid signaling. Further experiments have demonstrated cKO mice exhibit elevated uterine PGR signaling and reduced estrogen receptor signaling, although the levels of progesterone and estrogen remained unaltered. These alterations may contribute to defects in epithelium. Discussion Overall, our findings highlight a previously unrecognized but indispensable role for DCAF2 in the development of uterine luminal and glandular epithelium by orchestrating PGR and estrogen receptor responses. Its deficiency in the uterus leads to mouse infertility.
Collapse
Affiliation(s)
- Man Yang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Kaixuan Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Liang Zhang
- Research Center of Translational Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hongya Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
7
|
Dias Da Silva I, Wuidar V, Zielonka M, Pequeux C. Unraveling the Dynamics of Estrogen and Progesterone Signaling in the Endometrium: An Overview. Cells 2024; 13:1236. [PMID: 39120268 PMCID: PMC11312103 DOI: 10.3390/cells13151236] [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: 05/24/2024] [Revised: 06/25/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
The endometrium is crucial for the perpetuation of human species. It is a complex and dynamic tissue lining the inner wall of the uterus, regulated throughout a woman's life based on estrogen and progesterone fluctuations. During each menstrual cycle, this multicellular tissue undergoes cyclical changes, including regeneration, differentiation in order to allow egg implantation and embryo development, or shedding of the functional layer in the absence of pregnancy. The biology of the endometrium relies on paracrine interactions between epithelial and stromal cells involving complex signaling pathways that are modulated by the variations of estrogen and progesterone levels across the menstrual cycle. Understanding the complexity of estrogen and progesterone receptor signaling will help elucidate the mechanisms underlying normal reproductive physiology and provide fundamental knowledge contributing to a better understanding of the consequences of hormonal imbalances on gynecological conditions and tumorigenesis. In this narrative review, we delve into the physiology of the endometrium, encompassing the complex signaling pathways of estrogen and progesterone.
Collapse
Grants
- J.0165.24, 7.6529.23, J.0153.22, 7.4580.21F, 7.6518.21, J.0131.19 Fund for Scientific Research
- FSR-F-2023-FM, FSR-F-2022-FM, FSR-F-2021-FM, FSR-F-M-19/6761 University of Liège
- 2020, 2021, 2022 Fondation Léon Fredericq
Collapse
Affiliation(s)
| | | | | | - Christel Pequeux
- Tumors and Development, Estrogen-Sensitive Tissues and Cancer Team, GIGA-Cancer, Laboratory of Biology, University of Liège, 4000 Liège, Belgium; (I.D.D.S.); (V.W.); (M.Z.)
| |
Collapse
|
8
|
Kowsar R, Sadeghi K, Hashemzadeh F, Miyamoto A. Ovarian sex steroid and epithelial control of immune responses in the uterus and oviduct: human and animal models†. Biol Reprod 2024; 110:230-245. [PMID: 38038990 PMCID: PMC10873282 DOI: 10.1093/biolre/ioad166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/08/2023] [Accepted: 11/30/2023] [Indexed: 12/02/2023] Open
Abstract
The female reproductive tract (FRT), including the uterus and oviduct (Fallopian tube), is responsible for maintaining an optimal microenvironment for reproductive processes, such as gamete activation and transportation, sperm capacitation, fertilization, and early embryonic and fetal development. The mucosal surface of the FRT may be exposed to pathogens and sexually transmitted microorganisms due to the opening of the cervix during mating. Pathogens and endotoxins may also reach the oviduct through the peritoneal fluid. To maintain an optimum reproductive environment while recognizing and killing pathogenic bacterial and viral agents, the oviduct and uterus should be equipped with an efficient and rigorously controlled immune system. Ovarian sex steroids can affect epithelial cells and underlying stromal cells, which have been shown to mediate innate and adaptive immune responses. This, in turn, protects against potential infections while maintaining an optimal milieu for reproductive events, highlighting the homeostatic involvement of ovarian sex steroids and reproductive epithelial cells. This article will discuss how ovarian sex steroids affect the immune reactions elicited by the epithelial cells of the non-pregnant uterus and oviduct in the bovine, murine, and human species. Finally, we propose that there are regional and species-specific differences in the immune responses in FRT.
Collapse
Affiliation(s)
- Rasoul Kowsar
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | | | - Farzad Hashemzadeh
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Akio Miyamoto
- Global Agromedicine Research Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| |
Collapse
|
9
|
Colón-Caraballo M, Flores-Caldera I. Translational aspects of the endometriosis epigenome. EPIGENETICS IN HUMAN DISEASE 2024:883-929. [DOI: 10.1016/b978-0-443-21863-7.00008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Chen Z, Dean M. Endometrial Glucose Metabolism During Early Pregnancy. REPRODUCTION AND FERTILITY 2023; 4:RAF-23-0016. [PMID: 37934727 PMCID: PMC10762551 DOI: 10.1530/raf-23-0016] [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: 03/03/2023] [Accepted: 11/07/2023] [Indexed: 11/09/2023] Open
Abstract
Approximately 50% of human pregnancies humans fail, most before or during implantation. One factor contributing to pregnancy loss is abnormal glucose metabolism in the endometrium. Glucose contributes to preimplantation embryo development, uterine receptivity, and attachment of the embryo. Across multiple species, the epithelium stores glucose as the macromolecule glycogen at estrus. This reserve is mobilized during the preimplantation period. Glucose from circulation or glycogenolysis can be secreted into the uterine lumen for use by the embryo or metabolized via glycolysis, producing ATP for the cell. The resulting pyruvate could be converted to lactate, another important nutrient for the embryo. Fructose is an important nutrient for early embryos, and the epithelium and placenta can convert glucose to fructose via the polyol pathway. The epithelium also uses glucose to glycosylate proteins, which regulates embryo attachment. In some species, decidualization of the stroma is critical to successful implantation. Formation of the decidua requires increased glucose metabolism via the pentose phosphate pathway and glycolysis. After decidualization, the cells switch to aerobic glycolysis to produce ATP. Paradoxically, the decidua also stores large amounts of glucose as glycogen. Too little glucose or an inability to take up glucose impairs embryo development and decidualization. Conversely, too much glucose inhibits these same processes. This likely contributes to the reduced pregnancy rates associated with conditions like obesity and diabetes. Collectively, precise control of glucose metabolism is important for several endometrial processes required to establish a successful pregnancy. The factors regulating these metabolic processes remain poorly understood.
Collapse
Affiliation(s)
- Ziting Chen
- Department of Animal Science, University of Illinois, Urbana-Champaign, Urbana, Illinois, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew Dean
- Department of Animal Science, University of Illinois, Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
11
|
Rizo JA, Davenport KM, Winuthayanon W, Spencer TE, Kelleher AM. Estrogen receptor alpha regulates uterine epithelial lineage specification and homeostasis. iScience 2023; 26:107568. [PMID: 37622003 PMCID: PMC10445454 DOI: 10.1016/j.isci.2023.107568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/11/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
Postnatal development of the uterus involves specification of undifferentiated epithelium into uterine-type epithelium. That specification is regulated by stromal-epithelial interactions as well as intrinsic cell-specific transcription factors and gene regulatory networks. This study utilized mouse genetic models of Esr1 deletion, endometrial epithelial organoids (EEO), and organoid-stromal co-cultures to decipher the role of Esr1 in uterine epithelial development. Organoids derived from wild-type (WT) mice developed a normal single layer of columnar epithelium. In contrast, EEO from Esr1 null mice developed a multilayered stratified squamous type of epithelium with basal cells. Co-culturing Esr1 null epithelium with WT uterine stromal fibroblasts inhibited basal cell development. Of note, estrogen treatment of EEO-stromal co-cultures and Esr1 conditional knockout mice increased basal epithelial cell markers. Collectively, these findings suggest that Esr1 regulates uterine epithelium lineage plasticity and homeostasis and loss of ESR1 promotes altered luminal-to-basal differentiation driven by ESR1-mediated paracrine factors from the stroma.
Collapse
Affiliation(s)
- Jason A. Rizo
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | | | - Wipawee Winuthayanon
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| | - Thomas E. Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| | - Andrew M. Kelleher
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
12
|
Gnecco JS, Brown A, Buttrey K, Ives C, Goods BA, Baugh L, Hernandez-Gordillo V, Loring M, Isaacson KB, Griffith LG. Organoid co-culture model of the human endometrium in a fully synthetic extracellular matrix enables the study of epithelial-stromal crosstalk. MED 2023; 4:554-579.e9. [PMID: 37572651 PMCID: PMC10878405 DOI: 10.1016/j.medj.2023.07.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 03/11/2023] [Accepted: 07/11/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND The human endometrium undergoes recurring cycles of growth, differentiation, and breakdown in response to sex hormones. Dysregulation of epithelial-stromal communication during hormone-mediated signaling may be linked to myriad gynecological disorders for which treatments remain inadequate. Here, we describe a completely defined, synthetic extracellular matrix that enables co-culture of human endometrial epithelial and stromal cells in a manner that captures healthy and disease states across a simulated menstrual cycle. METHODS We parsed cycle-dependent endometrial integrin expression and matrix composition to define candidate cell-matrix interaction cues for inclusion in a polyethylene glycol (PEG)-based hydrogel crosslinked with matrix metalloproteinase-labile peptides. We semi-empirically screened a parameter space of biophysical and molecular features representative of the endometrium to define compositions suitable for hormone-driven expansion and differentiation of epithelial organoids, stromal cells, and co-cultures of the two cell types. FINDINGS Each cell type exhibited characteristic morphological and molecular responses to hormone changes when co-encapsulated in hydrogels tuned to a stiffness regime similar to the native tissue and functionalized with a collagen-derived adhesion peptide (GFOGER) and a fibronectin-derived peptide (PHSRN-K-RGD). Analysis of cell-cell crosstalk during interleukin 1B (IL1B)-induced inflammation revealed dysregulation of epithelial proliferation mediated by stromal cells. CONCLUSIONS Altogether, we demonstrate the development of a fully synthetic matrix to sustain the dynamic changes of the endometrial microenvironment and support its applications to understand menstrual health and endometriotic diseases. FUNDING This work was supported by The John and Karine Begg Foundation, the Manton Foundation, and NIH U01 (EB029132).
Collapse
Affiliation(s)
- Juan S Gnecco
- Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Alexander Brown
- Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Kira Buttrey
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Clara Ives
- Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Brittany A Goods
- Thayer School of Engineering at Dartmouth College, 15 Thayer Drive, Hanover, NH 03755, USA
| | - Lauren Baugh
- Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Victor Hernandez-Gordillo
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Megan Loring
- Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Newton Wellesley Hospital, 2014 Washington Street, Newton, MA 02115, USA
| | - Keith B Isaacson
- Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Newton Wellesley Hospital, 2014 Washington Street, Newton, MA 02115, USA
| | - Linda G Griffith
- Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
13
|
Furuminato K, Minatoya S, Senoo E, Goto T, Yamazaki S, Sakaguchi M, Toyota K, Iguchi T, Miyagawa S. The role of mesenchymal estrogen receptor 1 in mouse uterus in response to estrogen. Sci Rep 2023; 13:12293. [PMID: 37516793 PMCID: PMC10387046 DOI: 10.1038/s41598-023-39474-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023] Open
Abstract
Estrogens play important roles in uterine growth and homeostasis through estrogen receptors (ESR1 and ESR2). To address the role of ESR1-mediated tissue events in the murine uterus, we analyzed mice with a mesenchymal tissue-specific knockout of Esr1. Isl1-driven Cre expression generated Esr1 deletion in the uterine stroma and endometrium (Isl-Esr1KO). We showed that overall structure of the Isl1-Esr1KO mouse uterus developed normally, but estrogen responsiveness and subsequent growth were defective, suggesting that mesenchymal ESR1 is necessary for both epithelial and mesenchymal cell proliferation. Furthermore, RNA-seq analysis revealed that the majority of estrogen-induced genes were regulated by stromal ESR1. In control mice, E2 administration induced 9476 up-regulated differentially expressed genes (DEGs), whereas only 1801 up-regulated DEGs were induced by E2 in Isl1-Esr1KO mice. We further showed that stromal ESR1-regulated genes in the mouse uterus included several growth factors and cytokines, which are potential factors that regulate epithelial and stromal tissue interaction, and also genes involved in lipid homeostasis. Therefore, we infer that stromal ESR1 expression is indispensable for most estrogen actions in the mouse uterus and the current results provide new insights into estrogen-mediated homeostasis in female reproductive organs.
Collapse
Affiliation(s)
- Keita Furuminato
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Saki Minatoya
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Eriko Senoo
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Tatsuki Goto
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Sho Yamazaki
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Moeka Sakaguchi
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Kenji Toyota
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Noto, Ishikawa, 927-0552, Japan
| | - Taisen Iguchi
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa, 236-0027, Japan
| | - Shinichi Miyagawa
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan.
| |
Collapse
|
14
|
Fitzgerald HC, Kelleher AM, Ranjit C, Schust DJ, Spencer TE. Basolateral secretions of human endometrial epithelial organoids impact stromal cell decidualization. Mol Hum Reprod 2023; 29:gaad007. [PMID: 36821428 PMCID: PMC10321591 DOI: 10.1093/molehr/gaad007] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/19/2023] [Indexed: 02/24/2023] Open
Abstract
Uterine glands and, by inference, their secretions impact uterine receptivity, blastocyst implantation, stromal cell decidualization, and placental development. Changes in gland function across the menstrual cycle are primarily governed by the steroid hormones estrogen (E2) and progesterone (P4) but can also be influenced by extrinsic factors from the stroma. Using a human endometrial epithelial organoid system, transcriptome and proteome analyses identified distinct responses of the organoids to steroid hormones and prostaglandin E2 (PGE2). Notably, P4 and PGE2 modulated the basolateral secretion of organoid proteins, particularly cystatin C (CST3), serpin family A member 3 (SERPINA3), and stanniocalcin 1 (STC1). CST3, but not SERPINA3 or STC1, attenuated the in vitro stromal decidualization response to steroid hormones and PGE2. These findings provide evidence that uterine gland-derived factors impact stromal cell decidualization, which has implications for pregnancy establishment and fertility in women.
Collapse
Affiliation(s)
- Harriet C Fitzgerald
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Present address: The Ritchie Centre, Hudson Institute of Medical Research, Clayton, 3168 Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, 3168 Victoria, Australia
| | - Andrew M Kelleher
- Division of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO, USA
| | - Chaman Ranjit
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Danny J Schust
- Division of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO, USA
- Present address: Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Division of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO, USA
| |
Collapse
|
15
|
Zeng X, Li S, Liu L, Cai S, Ye Q, Xue B, Wang X, Zhang S, Chen F, Cai C, Wang F, Zeng X. Role of functional fatty acids in modulation of reproductive potential in livestock. J Anim Sci Biotechnol 2023; 14:24. [PMID: 36788613 PMCID: PMC9926833 DOI: 10.1186/s40104-022-00818-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/04/2022] [Indexed: 02/16/2023] Open
Abstract
Fatty acids are not only widely known as energy sources, but also play important roles in many metabolic pathways. The significance of fatty acids in modulating the reproductive potential of livestock has received greater recognition in recent years. Functional fatty acids and their metabolites improve follicular development, oocyte maturation and embryo development, as well as endometrial receptivity and placental vascular development, through enhancing energy supply and precursors for the synthesis of their productive hormones, such as steroid hormones and prostaglandins. However, many studies are focused on the impacts of individual functional fatty acids in the reproductive cycle, lacking studies involved in deeper mechanisms and optimal fatty acid requirements for specific physiological stages. Therefore, an overall consideration of the combination and synergy of functional fatty acids and the establishment of optimal fatty acid requirement for specific stages is needed to improve reproductive potential in livestock.
Collapse
Affiliation(s)
- Xiangzhou Zeng
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Siyu Li
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Lu Liu
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Shuang Cai
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Qianhong Ye
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, 430070 Wuhan, Hubei China
| | - Bangxin Xue
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Xinyu Wang
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Shihai Zhang
- grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Fang Chen
- grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Chuanjiang Cai
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, 712100 Yangling, Shaanxi China
| | - Fenglai Wang
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193, Beijing, P. R. China. .,Beijing Key Laboratory of Bio feed Additives, 100193, Beijing, P. R. China.
| |
Collapse
|
16
|
Devillers MM, Mhaouty-Kodja S, Guigon CJ. Deciphering the Roles & Regulation of Estradiol Signaling during Female Mini-Puberty: Insights from Mouse Models. Int J Mol Sci 2022; 23:ijms232213695. [PMID: 36430167 PMCID: PMC9693133 DOI: 10.3390/ijms232213695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Mini-puberty of infancy is a short developmental phase occurring in humans and other mammals after birth. In females, it corresponds to transient and robust activation of the hypothalamo-pituitary-ovarian (HPO) axis revealed by high levels of gonadotropin hormones, follicular growth, and increased estradiol production by the ovary. The roles of estradiol signaling during this intriguing developmental phase are not yet well known, but accumulating data support the idea that it aids in the implementation of reproductive function. This review aims to provide in-depth information on HPO activity during this particular developmental phase in several mammal species, including humans, and to propose emerging hypotheses on the putative effect of estradiol signaling on the development and function of organs involved in female reproduction.
Collapse
Affiliation(s)
- Marie M. Devillers
- Sorbonne Paris Cité, Université de Paris Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l’Axe Gonadotrope U1133, CEDEX 13, 75205 Paris, France
| | - Sakina Mhaouty-Kodja
- Neuroscience Paris Seine—Institut de Biologie Paris Seine, Sorbonne Université, CNRS UMR 8246, INSERM U1130, 75005 Paris, France
| | - Céline J. Guigon
- Sorbonne Paris Cité, Université de Paris Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l’Axe Gonadotrope U1133, CEDEX 13, 75205 Paris, France
- Correspondence:
| |
Collapse
|
17
|
Wan S, Sun Y, Fu J, Song H, Xiao Z, Yang Q, Wang S, Yu G, Feng P, Lv W, Luo L, Guan Z, Liu F, Zhou Q, Yin Z, Yang M. mTORC1 signaling pathway integrates estrogen and growth factor to coordinate vaginal epithelial cells proliferation and differentiation. Cell Death Dis 2022; 13:862. [PMID: 36220823 PMCID: PMC9553898 DOI: 10.1038/s41419-022-05293-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022]
Abstract
The mouse vaginal epithelium cyclically exhibits cell proliferation and differentiation in response to estrogen. Estrogen acts as an activator of mTOR signaling but its role in vaginal epithelial homeostasis is unknown. We analyzed reproductive tract-specific Rptor or Rictor conditional knockout mice to reveal the role of mTOR signaling in estrogen-dependent vaginal epithelial cell proliferation and differentiation. Loss of Rptor but not Rictor in the vagina resulted in an aberrant proliferation of epithelial cells and failure of keratinized differentiation. As gene expression analysis indicated, several estrogen-mediated genes, including Pgr and Ereg (EGF-like growth factor) were not induced by estrogen in Rptor cKO mouse vagina. Moreover, supplementation of EREG could activate the proliferation and survival of vaginal epithelial cells through YAP1 in the absence of Rptor. Thus, mTORC1 signaling integrates estrogen and growth factor signaling to mediate vaginal epithelial cell proliferation and differentiation, providing new insights into vaginal atrophy treatment for post-menopausal women.
Collapse
Affiliation(s)
- Shuo Wan
- grid.258164.c0000 0004 1790 3548The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632 China ,grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Yadong Sun
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Jiamin Fu
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Hongrui Song
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Zhiqiang Xiao
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Quanli Yang
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Sanfeng Wang
- grid.459579.30000 0004 0625 057XGuangdong Women and Children Hospital, Guangzhou, Guangdong 510010 China
| | - Gongwang Yu
- grid.12981.330000 0001 2360 039XDepartment of Medical Genetics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Peiran Feng
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Wenkai Lv
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Liang Luo
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Zerong Guan
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Feng Liu
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Qinghua Zhou
- grid.258164.c0000 0004 1790 3548The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632 China ,grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Zhinan Yin
- grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| | - Meixiang Yang
- grid.258164.c0000 0004 1790 3548The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632 China ,grid.258164.c0000 0004 1790 3548Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.258164.c0000 0004 1790 3548The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632 Guangdong China
| |
Collapse
|
18
|
Chávez-Genaro R, Toledo A, Hernández K, Anesetti G. Structural and functional changes in rat uterus induced by neonatal androgenization. J Mol Histol 2022; 53:903-914. [PMID: 36201133 DOI: 10.1007/s10735-022-10106-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022]
Abstract
Fetal or neonatal androgen exposure has a programming effect on ovarian function inducing a polycystic ovarian syndrome-like condition. Its effects on uterine structure and function are poorly studied. The aim of this work was to characterize the temporal course of changes in the rat uterine structure induced by neonatal exposure to aromatizable or not aromatizable androgens. Rats were daily treated with testosterone, dihydrotestosterone or vehicle during follicle assembly period (postnatal days 1 to 5). Uterine histoarchitecture, hormonal milieu, endometrial stromal collagen and capillary density were analyzed at prepubertal, pubertal and adult ages. Our data shows that neonatal androgen exposure induces early and long-lasting deleterious effects on uterine development, including altered adenogenesis and superficial epithelial alterations and suggest a role for altered serum estradiol levels in the maintenance and worsening of the situation. Our results suggest that alterations of the neonatal androgenic environment on the uterus could be responsible for alterations in the processes of implantation and maintenance of the embryo in women with polycystic ovary syndrome.
Collapse
Affiliation(s)
- Rebeca Chávez-Genaro
- Laboratorio de Biología de la Reproducción, Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Agustina Toledo
- Laboratorio de Biología de la Reproducción, Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Karina Hernández
- Laboratorio de Biología de la Reproducción, Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Gabriel Anesetti
- Laboratorio de Biología de la Reproducción, Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
19
|
Tsolova AO, Aguilar RM, Maybin JA, Critchley HOD. Pre-clinical models to study abnormal uterine bleeding (AUB). EBioMedicine 2022; 84:104238. [PMID: 36081283 PMCID: PMC9465267 DOI: 10.1016/j.ebiom.2022.104238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022] Open
Abstract
Abnormal Uterine Bleeding (AUB) is a common debilitating condition that significantly reduces quality of life of women across the reproductive age span. AUB creates significant morbidity, medical, social, and economic problems for women, their families, workplace, and health services. Despite the profoundly negative effects of AUB on public health, advancement in understanding the pathophysiology of AUB and the discovery of novel effective therapies is slow due to lack of reliable pre-clinical models. This review discusses currently available laboratory-based pre-clinical scientific models and how they are used to study AUB. Human and animal in vitro, ex vivo, and in vivo models will be described along with advantages and limitations of each method.
Collapse
|
20
|
Molecular Mechanism of Equine Endometrosis: The NF-κB-Dependent Pathway Underlies the Ovarian Steroid Receptors’ Dysfunction. Int J Mol Sci 2022; 23:ijms23137360. [PMID: 35806363 PMCID: PMC9266418 DOI: 10.3390/ijms23137360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 12/10/2022] Open
Abstract
Endometrosis is a frequently occurring disease decreasing mares’ fertility. Thus, it is an important disease of the endometrium associated with epithelial and stromal cell alterations, endometrium gland degeneration and periglandular fibrosis. Multiple degenerative changes are found in uterine mucosa, the endometrium. However, their pathogenesis is not well known. It is thought that nuclear factor-κB (NF-κB), a cell metabolism regulator, and its activation pathways take part in it. The transcription of the profibrotic pathway genes of the NF-κB in fibrotic endometria differed between the follicular (FLP) and mid-luteal (MLP) phases of the estrous cycle, as well as with fibrosis progression. This study aimed to investigate the transcription of genes of estrogen (ESR1, ESR2) and progesterone receptors (PGR) in equine endometria to find relationships between the endocrine environment, NF-κB-pathway, and fibrosis. Endometrial samples (n = 100), collected in FLP or MLP, were classified histologically, and examined using quantitative PCR. The phase of the cycle was determined through the evaluation of ovarian structures and hormone levels (estradiol, progesterone) in serum. The transcription of ESR1, ESR2, and PGR decreased with the severity of endometrial fibrosis and degeneration of the endometrium. Moreover, differences in the transcription of ESR1, ESR2, and PGR were noted between FLP and MLP in the specific categories and histopathological type of equine endometrosis. In FLP and MLP, specific moderate and strong correlations between ESR1, ESR2, PGR and genes of the NF-κB pathway were evidenced. The transcription of endometrial steroid receptors can be subjected to dysregulation with the degree of equine endometrosis, especially in both destructive types of endometrosis, and mediated by the canonical NF-κB pathway depending on the estrous cycle phase.
Collapse
|
21
|
Aikaterini B, Sophia Z, Fanourios M, Panagiotis D, Timur G, Antonios M. Aging, a modulator of human endometrial stromal cell proliferation and decidualization. A role for implantation? Reprod Biomed Online 2022; 45:202-210. [DOI: 10.1016/j.rbmo.2022.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 01/09/2023]
|
22
|
Li K, Diakite D, Austin J, Lee J, Lantvit DD, Murphy BT, Burdette JE. The Flavonoid Baicalein Negatively Regulates Progesterone Target Genes in the Uterus in Vivo. JOURNAL OF NATURAL PRODUCTS 2022; 85:237-247. [PMID: 34935393 PMCID: PMC9164990 DOI: 10.1021/acs.jnatprod.1c01008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Baicalein is a flavonoid extracted from the root of Scutellaria baicalensis (Chinese skullcap) and is consumed as part of this botanical dietary supplement to reduce oxidative stress, pain, and inflammation. We previously reported that baicalein can also modify receptor signaling through the progesterone receptor (PR) and glucocorticoid receptor (GR) in vitro, which is interesting due to the well-established roles of both PR and GR in reducing inflammation. To understand the effects of baicalein on PR and GR signaling in vivo in the uterus, ovariectomized CD-1 mice were treated with DMSO, progesterone (P4), baicalein, P4 with baicalein, and P4 with RU486, a PR antagonist, for a week. The uteri were collected for histology and RNA sequencing. Our results showed that baicalein attenuated the antiproliferative effect of P4 on luminal epithelium as well as on the PR target genes HAND2 and ZBTB16. Baicalein did not change levels of PR or GR RNA or protein in the uterus. RNA sequencing data indicated that many transcripts significantly altered by baicalein were regulated in the opposite direction by P4. Similarly, a large portion of GO/KEGG terms and GSEA gene sets were altered in the opposite direction by baicalein as compared to P4 treatment. Treatment of baicalein did not change body weight, organ weight, or blood glucose level. In summary, baicalein functioned as a PR antagonist in vivo and therefore may oppose P4 action under certain conditions such as uterine hyperplasia, fibroids, and uterine cancers.
Collapse
Affiliation(s)
- Kailiang Li
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Djeneba Diakite
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Julia Austin
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Jeongho Lee
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Daniel D. Lantvit
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Brian T. Murphy
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60607, USA
| |
Collapse
|
23
|
Berg MD, Chen Z, Dean M. Establishment and characterization of epithelial and fibroblast cell lines from the bovine endometrium. In Vitro Cell Dev Biol Anim 2022; 58:8-13. [DOI: 10.1007/s11626-021-00640-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/07/2021] [Indexed: 11/05/2022]
|
24
|
Farkas S, Szabó A, Török B, Sólyomvári C, Fazekas CL, Bánrévi K, Correia P, Chaves T, Zelena D. Ovariectomy-induced hormone deprivation aggravates Aβ 1-42 deposition in the basolateral amygdala and cholinergic fiber loss in the cortex but not cognitive behavioral symptoms in a triple transgenic mouse model of Alzheimer's disease. Front Endocrinol (Lausanne) 2022; 13:985424. [PMID: 36303870 PMCID: PMC9596151 DOI: 10.3389/fendo.2022.985424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease is the most common type of dementia, being highly prevalent in elderly women. The advanced progression may be due to decreased hormone synthesis during post-menopause as estradiol and progesterone both have neuroprotective potentials. We aimed to confirm that female hormone depletion aggravates the progression of dementia in a triple transgenic mouse model of Alzheimer's disease (3xTg-AD). As pathological hallmarks are known to appear in 6-month-old animals, we expected to see disease-like changes in the 4-month-old 3xTg-AD mice only after hormone depletion. Three-month-old female 3xTg-AD mice were compared with their age-matched controls. As a menopause model, ovaries were removed (OVX or Sham surgery). After 1-month recovery, the body composition of the animals was measured by an MRI scan. The cognitive and anxiety parameters were evaluated by different behavioral tests, modeling different aspects (Y-maze, Morris water maze, open-field, social discrimination, elevated plus maze, light-dark box, fox odor, operant conditioning, and conditioned fear test). At the end of the experiment, uterus was collected, amyloid-β accumulation, and the cholinergic system in the brain was examined by immunohistochemistry. The uterus weight decreased, and the body weight increased significantly in the OVX animals. The MRI data showed that the body weight change can be due to fat accumulation. Moreover, OVX increased anxiety in control, but decreased in 3xTg-AD animals, the later genotype being more anxious by default based on the anxiety z-score. In general, 3xTg-AD mice moved less. In relation to cognition, neither the 3xTg-AD genotype nor OVX surgery impaired learning and memory in general. Despite no progression of dementia-like behavior after OVX, at the histological level, OVX aggravated the amyloid-β plaque deposition in the basolateral amygdala and induced early cholinergic neuronal fiber loss in the somatosensory cortex of the transgenic animals. We confirmed that OVX induced menopausal symptoms. Removal of the sexual steroids aggravated the appearance of AD-related alterations in the brain without significantly affecting the behavior. Thus, the OVX in young, 3-month-old 3xTg-AD mice might be a suitable model for testing the effect of new treatment options on structural changes; however, to reveal any beneficial effect on behavior, a later time point might be needed.
Collapse
Affiliation(s)
- Szidónia Farkas
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Budapest, Hungary
| | - Adrienn Szabó
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Bibiána Török
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Budapest, Hungary
| | - Csenge Sólyomvári
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Csilla Lea Fazekas
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Krisztina Bánrévi
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Budapest, Hungary
| | - Pedro Correia
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Tiago Chaves
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Dóra Zelena
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, Budapest, Hungary
- *Correspondence: Dóra Zelena,
| |
Collapse
|
25
|
Sirohi VK, Medrano TI, Mesa AM, Kannan A, Bagchi IC, Cooke PS. Regulation of AKT Signaling in Mouse Uterus. Endocrinology 2022; 163:bqab233. [PMID: 34791100 PMCID: PMC8667855 DOI: 10.1210/endocr/bqab233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Indexed: 01/02/2023]
Abstract
17β-estradiol (E2) treatment of ovariectomized adult mice stimulates the uterine PI3K-AKT signaling pathway and epithelial proliferation through estrogen receptor 1 (ESR1). However, epithelial proliferation occurs independently of E2/ESR1 signaling in neonatal uteri. Similarly, estrogen-independent uterine epithelial proliferation is seen in adulthood in mice lacking Ezh2, critical for histone methylation, and in wild-type (WT) mice treated neonatally with estrogen. The role of AKT in estrogen-independent uterine epithelial proliferation was the focus of this study. Expression of the catalytically active phosphorylated form of AKT (p-AKT) and epithelial proliferation were high in estrogen receptor 1 knockout and WT mice at postnatal day 6, when E2 concentrations were low, indicating that neither ESR1 nor E2 are essential for p-AKT expression and epithelial proliferation in these mice. However, p-AKT levels and proliferation remained estrogen responsive in preweaning WT mice. Expression of p-AKT and proliferation were both high in uterine luminal epithelium of mice estrogenized neonatally and ovariectomized during adulthood. Increased expression of phosphorylated (inactive) EZH2 was also observed. Consistent with this, Ezh2 conditional knockout mice show ovary-independent uterine epithelial proliferation and high epithelial p-AKT. Thus, adult p-AKT expression is constitutive and E2/ESR1 independent in both model systems. Finally, E2-induced p-AKT expression and normal uterine proliferation did not occur in mice lacking membrane (m)ESR1, indicating a key role for membrane ESR1 in AKT activation. These findings emphasize the importance of AKT activation in promoting uterine epithelial proliferation even when that proliferation is not E2/ESR1 dependent and further indicate that p-AKT can be uncoupled from E2/ESR1 signaling in several experimental scenarios.
Collapse
Affiliation(s)
- Vijay K Sirohi
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Theresa I Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Athilakshmi Kannan
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Indrani C Bagchi
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
26
|
Insight on Polyunsaturated Fatty Acids in Endometrial Receptivity. Biomolecules 2021; 12:biom12010036. [PMID: 35053184 PMCID: PMC8773570 DOI: 10.3390/biom12010036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
Abstract
Endometrial receptivity plays a crucial role in fertilization as well as pregnancy outcome in patients faced with fertility challenges. The optimization of endometrial receptivity may help with normal implantation of the embryo, and endometrial receptivity may be affected by numerous factors. Recently, the role of lipids in pregnancy has been increasingly recognized. Fatty acids and their metabolites may be involved in all stages of pregnancy and play a role in supporting cell proliferation and development, participating in cell signaling and regulating cell function. Polyunsaturated fatty acids, in particular, are essential fatty acids for the human body that can affect the receptivity of the endometrium through in a variety of methods, such as producing prostaglandins, estrogen and progesterone, among others. Additionally, polyunsaturated fatty acids are also involved in immunity and the regulation of endometrial decidualization. Fatty acids are essential for fetal placental growth and development. The interrelationship of polyunsaturated fatty acids with these substances and how they may affect endometrial receptivity will be reviewed in this article.
Collapse
|
27
|
Abstract
Uniquely among adult tissues, the human endometrium undergoes cyclical shedding, scar-free repair and regeneration during a woman's reproductive life. Therefore, it presents an outstanding model for study of such processes. This Review examines what is known of endometrial repair and regeneration following menstruation and parturition, including comparisons with wound repair and the influence of menstrual fluid components. We also discuss the contribution of endometrial stem/progenitor cells to endometrial regeneration, including the importance of the stem cell niche and stem cell-derived extracellular vesicles. Finally, we comment on the value of endometrial epithelial organoids to extend our understanding of endometrial development and regeneration, as well as therapeutic applications.
Collapse
Affiliation(s)
- Lois A Salamonsen
- Centre for Reproductive Health, Clayton, Victoria 3168, Australia.,Department of Molecular and Translational Science, Clayton, Victoria 3168, Australia
| | - Jennifer C Hutchison
- Centre for Reproductive Health, Clayton, Victoria 3168, Australia.,Department of Molecular and Translational Science, Clayton, Victoria 3168, Australia
| | - Caroline E Gargett
- Ritchie Centre, Hudson Institute of Medical Research, 25-31 Wright St, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
28
|
Functional roles of female sex hormones and their nuclear receptors in cervical cancer. Essays Biochem 2021; 65:941-950. [PMID: 34156060 DOI: 10.1042/ebc20200175] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/21/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022]
Abstract
There has been little progress for several decades in modalities to treat cervical cancer. While the cervix is a hormone-sensitive tissue, physiologic roles of estrogen receptor α (ERα), progesterone receptor (PR), and their ligands in this tissue are poorly understood. It has hampered critical assessments of data in early epidemiologic and clinical studies for cervical cancer. Experimental evidence obtained from studies using mouse models has provided new insights into the molecular mechanism of ERα and PR in cervical cancer. In a mouse model expressing human papillomavirus (HPV) oncogenes, exogenous estrogen promotes cervical cancer through stromal ERα. In the same mouse model, genetic ablation of PR promotes cervical carcinogenesis without exogenous estrogen. Medroxyprogesterone acetate, a PR-activating drug, regresses cervical cancer in the mouse model. These results support that ERα and PR play opposite roles in cervical cancer. They further support that ERα inhibition and PR activation may be translated into valuable treatment for a subset of cervical cancers.
Collapse
|
29
|
Simão VA, Lupi Júnior LA, Adan Araujo Leite G, Cherici Camargo IC, de Almeida Chuffa LG. Nandrolone decanoate causes uterine injury by changing hormone levels and sex steroid receptors in a dose- and time-dependent manner. Reprod Toxicol 2021; 102:98-108. [PMID: 33984419 DOI: 10.1016/j.reprotox.2021.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/13/2021] [Accepted: 05/06/2021] [Indexed: 01/13/2023]
Abstract
Different doses of nandrolone decanoate (ND) were used to investigate the expression of uterine sex steroid receptors (AR, ER-α, and ER-β) and the levels of serum sex hormones after treatment and recovery periods in adult rats. ND doses of 1.87, 3.75, 7.5, or 15 mg/kg b.w. or mineral oil (control group) were injected subcutaneously for 15 days, and the experimental groups were divided into three periods of evaluation: (a) ND treatment for 15 days, (b) ND treatment followed by 30-day-recovery and (c) ND treatment followed by 60-day-recovery. Estrous cycle was monitored daily. At the end of each experimental period, rats were euthanized for the collection of serum samples and uterine tissues. All animals showed persistent diestrus and only the highest ND dose was capable of inducing persistent diestrus until 60-day-recovery. Immunoexpression of uterine sex steroid receptors varied in a time-dependent manner. While AR expression was increase after treatment period, ER-α and ER-β expressions decreased after 60- and 30-day-recovery, respectively. ND also increased the serum levels of testosterone, 17β-estradiol, and dihydrotestosterone, especially at the highest doses of 7.5 and 15 mg ND/kg until 30 days of recovery. The levels of progesterone were significantly reduced in all ND-treated animals. No significant difference was observed in the levels of follicle-stimulating hormone, whereas the levels of luteinizing hormone varied according to specific dose and period. We conclude that uterine sex steroid receptors and sex hormones are affected by ND administration and these alterations can be only restored following lower doses and long recovery periods.
Collapse
Affiliation(s)
- Vinícius Augusto Simão
- Department of Biotechnology, Faculty of Sciences and Letters of Assis, FCL/UNESP, SP, 19806-900, Brazil.
| | - Luiz Antonio Lupi Júnior
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, IBB/UNESP, SP, 16618-689, Brazil.
| | - Gabriel Adan Araujo Leite
- Department of Cell Biology, Embryology and Genetics, Biological Sciences Center, UFSC, SC, 88040-900, Brazil.
| | | | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, IBB/UNESP, SP, 16618-689, Brazil.
| |
Collapse
|
30
|
Mackens S, Santos-Ribeiro S, Racca A, Daneels D, Koch A, Essahib W, Verpoest W, Bourgain C, Van Riet I, Tournaye H, Brosens JJ, Lee YH, Blockeel C, Van de Velde H. The proliferative phase endometrium in IVF/ICSI: an in-cycle molecular analysis predictive of the outcome following fresh embryo transfer. Hum Reprod 2021; 35:130-144. [PMID: 31916571 DOI: 10.1093/humrep/dez218] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Does an early proliferative phase endometrial biopsy harvested during ovarian stimulation harbour information predictive of the outcome following fresh embryo transfer (ET) in that same cycle? SUMMARY ANSWER Transcriptome analysis of the whole-tissue endometrium did not reveal significant differential gene expression (DGE) in relation to the outcome; however, the secretome profile of isolated, cultured and in vitro decidualized endometrial stromal cells (EnSCs) varied significantly between patients who had a live birth compared to those with an implantation failure following fresh ET in the same cycle as the biopsy. WHAT IS KNOWN ALREADY In the majority of endometrial receptivity research protocols, biopsies are harvested during the window of implantation (WOI). This, however, precludes ET in that same cycle, which is preferable as the endometrium has been shown to adapt over time. Endometrial biopsies taken during ovarian stimulation have been reported not to harm the chances of implantation, and in such biopsies DGE has been observed between women who achieve pregnancy versus those who do not. The impact of the endometrial proliferative phase on human embryo implantation remains unclear, but deserves further attention, especially since in luteal phase endometrial biopsies, a transcriptional signature predictive for repeated implantation failure has been associated with reduced cell proliferation, possibly indicating proliferative phase involvement. Isolation, culture and in vitro decidualization (IVD) of EnSCs is a frequently applied basic research technique to assess endometrial functioning, and a disordered EnSC secretome has previously been linked with failed implantation. STUDY DESIGN, SIZE, DURATION This study was nested in a randomized controlled trial (RCT) investigating the effect of endometrial scratching during the early follicular phase of ovarian stimulation on clinical pregnancy rates after IVF/ICSI. Of the 96 endometrial biopsies available, after eliminating those without fresh ET and after extensive matching in order to minimize the risk of potential confounding, 18 samples were retained to study two clinical groups: nine biopsies of patients with a live birth versus nine biopsies of patients with an implantation failure, both following fresh ET performed in the same cycle as the biopsy. We studied the proliferative endometrium by analysing its transcriptome and by isolating, culturing and decidualizing EnSCs in vitro. We applied this latter technique for the first time on proliferative endometrial biopsies obtained during ovarian stimulation for in-cycle outcome prediction, in an attempt to overcome inter-cycle variability. PARTICIPANTS/MATERIALS, SETTING, METHODS RNA-sequencing was performed for 18 individual whole-tissue endometrial biopsies on an Illumina HiSeq1500 machine. DGE was analysed three times using different approaches (DESeq2, EdgeR and the Wilcoxon rank-sum test, all in R). EnSC isolation and IVD was performed (for 2 and 4 days) for a subset of nine samples, after which media from undifferentiated and decidualized cultures were harvested, stored at -80°C and later assayed for 45 cytokines using a multiplex suspension bead immunoassay. The analysis was performed by partial least squares regression modelling. MAIN RESULTS AND THE ROLE OF CHANCE After correction for multiple hypothesis testing, DGE analysis revealed no significant differences between endometrial samples from patients who had a live birth and those with an implantation failure following fresh ET. However secretome analysis after EnSC isolation and culture, showed two distinct clusters that clearly corresponded to the two clinical groups. Upon IVD, the secretome profiles shifted from that of undifferentiated cells but the difference between the two clinical groups remained yet were muted, suggesting convergence of cytokine profiles after decidualization. LIMITATIONS, REASONS FOR CAUTION Caution is warranted due to the limited sample size of the study and the in vitro nature of the EnSC experiment. Validation on a larger scale is necessary, however, hard to fulfil given the very limited availability of in-cycle proliferative endometrial biopsies outside a RCT setting. WIDER IMPLICATIONS OF THE FINDINGS These data support the hypothesis that the endometrium should be assessed not only during the WOI and that certain endometrial dysfunctionalities can probably be detected early in a cycle by making use of the proliferative phase. This insight opens new horizons for the development of endometrial tests, whether diagnostic or predictive of IVF/ICSI treatment outcome. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by Fonds Wetenschappelijk Onderzoek (FWO, Flanders, Belgium, 11M9415N, 1 524 417N), Wetenschappelijk Fonds Willy Gepts (WFWG G160, Universitair Ziekenhuis Brussel, Belgium) and the National Medicine Research Council (NMRC/CG/M003/2017, Singapore). There are no conflicts of interests. TRIAL REGISTRATION NUMBER NCT02061228.
Collapse
Affiliation(s)
- S Mackens
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - S Santos-Ribeiro
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,IVI-RMA Lisboa, Avenida Infante Dom Henrique 333 H 1-9, 1800-282 Lisbon, Portugal
| | - A Racca
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - D Daneels
- Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - A Koch
- Department of Pathology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - W Essahib
- Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - W Verpoest
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - C Bourgain
- Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Department of Pathology, Imelda Ziekenhuis Bonheiden, Bonheiden, Belgium
| | - I Van Riet
- Department of Hematology and Immunology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - H Tournaye
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - J J Brosens
- Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, UK
| | - Y H Lee
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore.,Obstetrics & Gynaecology-Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - C Blockeel
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - H Van de Velde
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Research group Reproduction and Immunology (REIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
31
|
Liu F, Li Z, Guo J, Fang S, Zhou J, Cao B, Liu J, Yi Y, Yuan X, Xu X, Huang O, Wang L, Zou Y. Endometrial stromal cell proteomic analysis reveals LIM and SH3 protein 1 (LASP1) plays important roles in the progression of adenomyosis. Mol Hum Reprod 2021; 27:6129094. [PMID: 33543750 DOI: 10.1093/molehr/gaab008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 01/12/2021] [Indexed: 11/14/2022] Open
Abstract
Adenomyosis is one of the most common gynecological disorders that the molecular events underlying its pathogenesis remain not fully understood. Prior studies have shown that endometrial stromal cells (ESCs) played crucial roles in the pathogenesis of adenomyosis. In this study, we utilized two-dimensional gel electrophoresis combined with protein identification by mass spectrometry (2D/MS) proteomics analysis to compare the differential protein expression profile between the paired eutopic and ectopic ESCs (EuESCs and EcESCs) in adenomyosis, and a total of 32 significantly altered protein spots were identified. Among which, the expression of LIM and SH3 protein 1 (LASP1) was increased significantly in EcESCs compared to EuESCs. Immunohistochemical assay showed that LASP1 was overexpressed in the stromal cells of ectopic endometriums compared to eutopic endometriums; further functional analyses revealed that LASP1 overexpression could enhance cell proliferation, migration and invasion of EcESCs. Furthermore, we also showed that the dysregulated expression of LASP1 in EcESCs was associated with DNA hypermethylation in the promoter region of the LASP1 gene. However, the detailed molecular mechanisms of enhancing cell proliferation, invasion and migration caused by upregulated LASP1 in adenomyosis needs further study. For the first time, our data suggested that LASP1 plays important roles in the pathogenesis of adenomyosis, and could serve as a prognostic biomarker of adenomyosis.
Collapse
Affiliation(s)
- Faying Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Zengming Li
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Jiubai Guo
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Shufen Fang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Jiangyan Zhou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Bianna Cao
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Jun Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Yulan Yi
- Department of Gynecology, Huangshi Central Hospital, Huangshi, Hubei, China
| | - Xiaoqun Yuan
- Department of Gynecology, Jiujiang Maternal and Child Health Hospital, Jiujiang, Jiangxi, China
| | - Xiaoyun Xu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Ouping Huang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Liqun Wang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Department of Reproductive Health, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
32
|
New Insights into Development of Female Reproductive Tract-Hedgehog-Signal Response in Wolffian Tissues Directly Contributes to Uterus Development. Int J Mol Sci 2021; 22:ijms22031211. [PMID: 33530552 PMCID: PMC7865753 DOI: 10.3390/ijms22031211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
The reproductive tract in mammals emerges from two ductal systems during embryogenesis: Wolffian ducts (WDs) and Mullerian ducts (MDs). Most of the female reproductive tract (FRT) including the oviducts, uterine horn and cervix, originate from MDs. It is widely accepted that the formation of MDs depends on the preformed WDs within the urogenital primordia. Here, we found that the WD mesenchyme under the regulation of Hedgehog (Hh) signaling is closely related to the developmental processes of the FRT during embryonic and postnatal periods. Deficiency of Sonic hedgehog (Shh), the only Hh ligand expressed exclusively in WDs, prevents the MD mesenchyme from affecting uterine growth along the radial axis. The in vivo cell tracking approach revealed that after WD regression, distinct cells responding to WD-derived Hh signal continue to exist in the developing FRT and gradually contribute to the formation of various tissues such as smooth muscle, endometrial stroma and vascular vessel, in the mouse uterus. Our study thus provides a novel developmental mechanism of FRT relying on WD.
Collapse
|
33
|
Beguelini MR, Santiago CS, Guerra LHA, Santos FCA, Góes RM, Morielle-Versute E, Taboga SR. The hormonal control of the uterus of the bat Myotis nigricans during its different reproductive phases: emphasis on progesterone and estradiol. Cell Tissue Res 2021; 384:211-229. [PMID: 33409655 DOI: 10.1007/s00441-020-03342-2] [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: 06/30/2020] [Accepted: 11/05/2020] [Indexed: 10/22/2022]
Abstract
Myotis nigricans is a species of bat from the Vespertilionidae family that is endemic of the Neotropical region. Its insectivorous feeding habit plus its large range of prey species, great geographical dispersion, wide colonies, and anthropomorphized behavior make this species an important ecological agent that acts in the control of nocturnal insects. Reproductively, M. nigricans presents geographic variations, having different patterns of reproduction according to its geographical location. Despite these extremely interesting characteristics, no more detailed study of the hormonal control of the reproduction of this species has been conducted. Therefore, the aim of the present study was to evaluate the variations in serum hormone concentrations and in uterine hormonal control of this bat during its different reproductive phases. Twenty adult females were collected, divided into four (4) sample groups, according to the reproductive status (nonreproductive, initial, and advanced pregnancy and lactating), and submitted to hormone dosage and immunohistochemical analyses. The results demonstrated that the uterus of M. nigricans is strongly regulated by the interaction/cross-talk between serum concentrations of estradiol (E2) and progesterone with their respective hormone receptors. Significant increases in the concentration of E2 and progesterone are needed to regulate the early pregnancy. The persistence of the corpus luteum throughout pregnancy is necessary, since its placenta does not express aromatase. The expressions of ERα and PR appear to be synchronized in order to coordinate a large portion of the processes that occur inside the uterus of M. nigricans during pregnancy and lactation.
Collapse
Affiliation(s)
- Mateus R Beguelini
- Center of Biological and Health Science, UFOB - Universidade Federal Do Oeste da Bahia, Barreiras, Bahia, Brazil.
| | - Cornélio S Santiago
- Center of Biological and Health Science, UFOB - Universidade Federal Do Oeste da Bahia, Barreiras, Bahia, Brazil
| | - Luiz H A Guerra
- Department of Biology, UNESP - University Estadual Paulista, São José Do Rio Preto, São Paulo, Brazil
| | - Fernanda C A Santos
- Department of Histology and Embryology, UFG - Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Rejane M Góes
- Department of Biology, UNESP - University Estadual Paulista, São José Do Rio Preto, São Paulo, Brazil
| | - Eliana Morielle-Versute
- Department of Zoology and Botany, UNESP - University Estadual Paulista, São José Do Rio Preto, São Paulo, Brazil
| | - Sebastião R Taboga
- Department of Biology, UNESP - University Estadual Paulista, São José Do Rio Preto, São Paulo, Brazil
| |
Collapse
|
34
|
Cunha GR, Li Y, Mei C, Derpinghaus A, Baskin LS. Ontogeny of estrogen receptors in human male and female fetal reproductive tracts. Differentiation 2020; 118:107-131. [PMID: 33176961 DOI: 10.1016/j.diff.2020.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/27/2022]
Abstract
This paper reviews and provides new observations on the ontogeny of estrogen receptor alpha (ESR1) and estrogen receptor beta (ESR2) in developing human male and female internal and external genitalia. Included in this study are observations on the human fetal uterine tube, the uterotubal junction, uterus, cervix, vagina, penis and clitoris. We also summarize and report on the ontogeny of estrogen receptors in the human fetal prostate, prostatic urethra and epididymis. The ontogeny of ESR1 and ESR2, which spans from 8 to 21 weeks correlates well with the known "window of susceptibility" (7-15 weeks) for diethylstilbestrol (DES)-induced malformations of the human female reproductive tract as determined through examination of DES daughters exposed in utero to this potent estrogen. Our fairly complete mapping of the ontogeny of ESR1 and ESR2 in developing human male and female internal and external genitalia provides a mechanistic framework for further investigation of the role of estrogen in normal development and of abnormalities elicited by exogenous estrogens.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | - Yi Li
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Cao Mei
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
35
|
Fitzgerald HC, Schust DJ, Spencer TE. In vitro models of the human endometrium: evolution and application for women's health. Biol Reprod 2020; 104:282-293. [PMID: 33009568 DOI: 10.1093/biolre/ioaa183] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
The endometrium is the inner lining of the uterus that undergoes complex regeneration and differentiation during the human menstrual cycle. The process of endometrial shedding, regeneration, and differentiation is driven by ovarian steroid hormones and prepares the endometrium and intrauterine environment for embryo implantation and pregnancy establishment. Endometrial glands and their secretions are essential for pregnancy establishment, and cross talk between the glandular epithelium and stromal cells appears vital for decidualization and placental development. Despite being crucial, the biology of the human endometrium during pregnancy establishment and most of pregnancy is incomplete, given the ethical and practical limitations of obtaining and studying endometrium from pregnant women. As such, in vitro models of the human endometrium are required to fill significant gaps in understanding endometrial biology. This review is focused on the evolution and development of in vitro three-dimensional models of the human endometrium and provides insight into the challenges and promises of those models to improve women's reproductive health.
Collapse
Affiliation(s)
| | - Danny J Schust
- Division of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA.,Division of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| |
Collapse
|
36
|
Dean M. Glycogen in the uterus and fallopian tubes is an important source of glucose during early pregnancy†. Biol Reprod 2020; 101:297-305. [PMID: 31201425 DOI: 10.1093/biolre/ioz102] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/16/2019] [Accepted: 06/06/2019] [Indexed: 01/02/2023] Open
Abstract
Pregnancy loss is common during the peri-implantation period in mammals when glucose is required for both embryonic development and decidualization of the endometrium. As the uterus cannot synthesize glucose, all glucose must come directly from maternal circulation as needed or transiently stored as the macromolecule glycogen. Glycogen acts as a glucose reservoir, storing up to 55 000 glucose moieties per molecule. Endometrial glycogen concentrations are correlated with fertility in humans, indicating that glycogen is an essential source of glucose during early pregnancy. In humans and primates, endometrial glycogen concentrations peak during the luteal phase due to progesterone. In contrast, in rats and mink, estradiol triggers an accumulation of uterine glycogen during proestrus and estrus. In mated rats, the glycogen content of the endometrium increases again after implantation due to high levels of glycogen stored in the decidua. In mink, endometrial glycogen reserves are localized in the uterine epithelia at estrus. These reserves are mobilized before implantation, suggesting they are used to support embryonic growth. Uterine glycogen concentrations continue to decrease after implantation in mink, probably due to a lack of decidualization. How ovarian steroids stimulate glycogenesis in the endometrium is unclear, but current evidence suggests that estradiol/progesterone interacts with insulin or insulin-like growth factor signaling. In summary, endometrial glycogen is an essential source of glucose during the peri-implantation period. More work is needed to characterize differences among species, elucidate the fate of the glucose liberated from glycogen, and understand how ovarian steroids regulate glycogen metabolism in the uterus.
Collapse
Affiliation(s)
- Matthew Dean
- Department of Animal Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
37
|
Wu J, Tao X, Zhang H, Yi XH, Yu YH. Estrogen-Induced Stromal FGF18 Promotes Proliferation and Invasion of Endometrial Carcinoma Cells Through ERK and Akt Signaling. Cancer Manag Res 2020; 12:6767-6777. [PMID: 32801905 PMCID: PMC7414926 DOI: 10.2147/cmar.s254242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Objective The aim of this study was to evaluate whether estrogen promoted the proliferation and invasion of endometrial carcinoma (EC) cells through paracrine FGFs in endometrial stromal cells (ESCs). Patients and Methods We screened gene alterations in a primary ESC culture after 10 nM estrogen treatment using an Agilent mRNA microarray. We knocked down stromal FGF18 expression in a co-culture system and aimed to explore the contribution of E2-induced stromal FGF18 to the proliferation and invasion of EC cells. To determine the effective receptors and detailed downstream signaling of FGF18, we co-cultured estrogen-treated hESCs with FGFR1-, FGFR2-, FGFR3- or FGFR4-knockdown Ishikawa cells. Finally, we detected FGF18 expression in clinical samples, including several primary cultures of different ESCs and a series of tissue microarrays (TMAs) of 90 patients with EC. Results A few genes altered significantly in estrogen-treated primary ESCs, but only FGF18 was noticeably enhanced among the FGF family genes. Knockdown of FGF18 expression in hESCs inhibited the promoting effect of FGF18 on the proliferation and invasion of EC cells. FGF18 bound FGFR2 and FGFR3 in Ishikawa cells to activate downstream ERK and Akt pathways and to promote the viability of EC cells. The FGF18-FGFR2 and FGF18-FGFR3 pathways had close correlations with Survivin and CD44V6 expression but not with P53. Primary ESCs of endometrioid EC (EEC, type I EC) had higher FGF18 expression than ESCs of normal endometrium (NE), endometrial atypical hyperplasia (EAH) and type II EC. Conclusion Estrogen induced FGF18 in ESCs to promote the proliferation and invasion of EC cells, and FGFR inhibitors should be considered as promising candidate targets for EC treatment.
Collapse
Affiliation(s)
- Jian Wu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, People's Republic of China.,Department of Pathology, Gongli Hospital, Second Military Medical University, Shanghai 200135, People's Republic of China
| | - Xiang Tao
- Department of Pathology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, People's Republic of China
| | - Hong Zhang
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, People's Republic of China
| | - Xiang-Hua Yi
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, People's Republic of China
| | - Yin-Hua Yu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, People's Republic of China
| |
Collapse
|
38
|
Hewitt SC, Carmona M, Foley KG, Donoghue LJ, Lierz SL, Winuthayanon W, Korach KS. Peri- and Postpubertal Estrogen Exposures of Female Mice Optimize Uterine Responses Later in Life. Endocrinology 2020; 161:bqaa081. [PMID: 32623449 PMCID: PMC7417879 DOI: 10.1210/endocr/bqaa081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/14/2020] [Indexed: 01/12/2023]
Abstract
At birth, all female mice, including those that either lack estrogen receptor α (ERα-knockout) or that express mutated forms of ERα (AF2ERKI), have a hypoplastic uterus. However, uterine growth and development that normally accompany pubertal maturation does not occur in ERα-knockout or AF2ERKI mice, indicating ERα-mediated estrogen (E2) signaling is essential for this process. Mice that lack Cyp19 (aromatase knockout, ArKO mice), an enzyme critical for E2 synthesis, are unable to make E2 and lack pubertal uterine development. A single injection of E2 into ovariectomized adult (10 weeks old) females normally results in uterine epithelial cell proliferation; however, we observe that although ERα is present in the ArKO uterine cells, no proliferative response is seen. We assessed the impact of exposing ArKO mice to E2 during pubertal and postpubertal windows and observed that E2-exposed ArKO mice acquired growth responsiveness. Analysis of differential gene expression between unexposed ArKO samples and samples from animals exhibiting the ability to mount an E2-induced uterine growth response (wild-type [WT] or E2-exposed ArKO) revealed activation of enhancer of zeste homolog 2 (EZH2) and heart- and neural crest derivatives-expressed protein 2 (HAND2) signaling and inhibition of GLI Family Zinc Finger 1 (GLI1) responses. EZH2 and HAND2 are known to inhibit uterine growth, and GLI1 is involved in Indian hedgehog signaling, which is a positive mediator of uterine response. Finally, we show that exposure of ArKO females to dietary phytoestrogens results in their acquisition of uterine growth competence. Altogether, our findings suggest that pubertal levels of endogenous and exogenous estrogens impact biological function of uterine cells later in life via ERα-dependent mechanisms.
Collapse
Affiliation(s)
- Sylvia C Hewitt
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Marleny Carmona
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - K Grace Foley
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Lauren J Donoghue
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Sydney L Lierz
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Wipawee Winuthayanon
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| | - Kenneth S Korach
- Reproductive and Development Biology Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina
| |
Collapse
|
39
|
Pluripotent Stem (VSELs) and Progenitor (EnSCs) Cells Exist in Adult Mouse Uterus and Show Cyclic Changes Across Estrus Cycle. Reprod Sci 2020; 28:278-290. [PMID: 32710237 DOI: 10.1007/s43032-020-00250-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/11/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022]
Abstract
We have earlier reported pluripotent, very small embryonic-like stem cells (VSELs) and slightly bigger endometrial stem cells (EnSCs) in adult mouse uterus and their regulation by gonadotropin and steroid hormones. VSELs can differentiate into cells of all three lineages in vitro; however, they neither expand readily in vitro nor compliment a developing embryo. In the present study, a robust protocol is described to enrich uterine stem/progenitor cells along with their characterization and variation across estrus cycle. After enzymatic digestion of adult mouse uterus, single-cell suspension obtained was spun at 1000 rpm (250 g) to pellet majority of cells. Stem cells remain buoyant at this speed and were pelleted by spinning supernatant at 3000 rpm (1000 g). Spherical, darkly stained VSELs (2-6 μm) with high nucleo-cytoplasmic ratio and EnSCs (> 6 μm) expressed OCT-4, NANOG, SSEA-1, SCA-1, and c-KIT. OCT-4-positive cells co-expressed SSEA-1, ERα, ERβ, PR, and FSHR. Transcripts specific for pluripotent state (Oct-4, Oct-4a, Sox-2, Nanog), primordial germ cells (Stella, Fragilis), and receptors for pituitary and steroid hormones (ERα, ERβ, PR, FSHR 1 and 3) were studied by RT-PCR in 3000 rpm pellet. Cell pellet collected at 3000 rpm showed 10-fold enrichment of VSELs (2-6 μm, viable cells with surface phenotype of LIN-CD45-SCA-1+) by flow cytometry and upregulation of pluripotent transcripts by qRT-PCR compared with 1000 rpm pellet. VSELs were maximal during estrus and metestrus phases of estrus cycle. To conclude, VSELs/EnSCs can be enriched from adult uterus using the strategy described here, vary in numbers across estrus cycle, and are vulnerable to endocrine disruption as they express steroid receptors.
Collapse
|
40
|
Zheng JH, Zhang JK, Kong DS, Song YB, Zhao SD, Qi WB, Li YN, Zhang ML, Huang XH. Quantification of the CM-Dil-labeled human umbilical cord mesenchymal stem cells migrated to the dual injured uterus in SD rat. Stem Cell Res Ther 2020; 11:280. [PMID: 32660551 PMCID: PMC7359016 DOI: 10.1186/s13287-020-01806-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 05/29/2020] [Accepted: 07/01/2020] [Indexed: 12/23/2022] Open
Abstract
Background Human umbilical cord mesenchymal stem cell (hUC-MSC) therapy is considered as a promising approach in the treatment of intrauterine adhesions (IUAs). Considerable researches have already detected hUC-MSCs by diverse methods. This paper aims at exploring the quantitative distribution of CM-Dil-labeled hUC-MSCs in different regions of the uterus tissue of the dual injury-induced IUAs in rats and the underlying mechanism of restoration of fertility after implantation of hUC-MSCs in the IUA model. Methods In this study, we investigated the quantification of the CM-Dil-labeled hUC-MSCs migrated to the dual injured uterus in Sprague Dawley rats. Additionally, we investigated the differentiation of CM-Dil-labeled hUC-MSCs. The differentiation potential of epithelial cells, vascular endothelial cells, and estrogen receptor (ER) cells were assessed by an immunofluorescence method using CK7, CD31, and ERα. The therapeutic impact of hUC-MSCs in the IUA model was assessed by hematoxylin and eosin, Masson, immunohistochemistry staining, and reproductive function test. Finally, the expression of TGF-β1/Smad3 pathway in uterine tissues was determined by qRT-PCR and Western blotting. Results The CM-Dil-labeled cells in the stroma region were significantly higher than those in the superficial myometrium (SM) (71.67 ± 7.98 vs. 60.92 ± 3.96, p = 0.005), in the seroma (71.67 ± 7.98 vs. 23.67 ± 8.08, p = 0.000) and in the epithelium (71.67 ± 7.98 vs. 4.17 ± 1.19, p = 0.000). From the 2nd week of treatment, hUC-MSCs began to differentiate into epithelial cells, vascular endothelial cells, and ER cells. The therapeutic group treated with hUC-MSCs exhibited a significant decrease in fibrosis (TGF-β1/Smad3) as well as a significant increase in vascularization (CD31) compared with the untreated rats. Conclusion Our findings suggested that the distribution of the migrated hUC-MSCs in different regions of the uterine tissue was unequal. Most cells were in the stroma and less were in the epithelium of endometrium and gland. Injected hUC-MSCs had a capacity to differentiate into epithelial cells, vascular endothelial cells, and ER cells; increase blood supply; inhibit fibration; and then restore the fertility of the IUA model.
Collapse
Affiliation(s)
- Jia-Hua Zheng
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing-Kun Zhang
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - De-Sheng Kong
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan-Biao Song
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shuang-Dan Zhao
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wen-Bo Qi
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ya-Nan Li
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ming-le Zhang
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiang-Hua Huang
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
41
|
Schöniger S, Schoon HA. The Healthy and Diseased Equine Endometrium: A Review of Morphological Features and Molecular Analyses. Animals (Basel) 2020; 10:ani10040625. [PMID: 32260515 PMCID: PMC7222714 DOI: 10.3390/ani10040625] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Diseases of the endometrium are a frequent cause of subfertility in mares and have an economic impact on the horse breeding industry. These include periglandular fibrosis of endometrial glands (endometrosis), degenerative diseases of vessels (angiosis), inflammation (endometritis), as well as altered differentiation of endometrial glands. Some mares are susceptible towards persistent endometritis. The etiology and pathogenesis of endometrosis are still unclear. This review describes morphological hallmarks and molecular features associated with endometrial health and different types of diseases. The presented literature data reveal characteristic differences in the expression of several extra- and intracellular molecules between the healthy and diseased equine endometrium. Some of these molecules can be detected directly within the tissue and thus have the potential to serve as excellent diagnostic markers for the presence of endometrial diseases. The knowledge of disease-associated changes in cellular differentiation, secretory functions, and immune mechanisms will help to decipher pathogenesis and will contribute to the development of novel treatments. In addition, the quantification of molecular alterations may contribute to a fertility prognosis for an individual mare. Reproductive health increases the well-being of mares and reduces financial loss for the horse breeding industry. Abstract Mares are seasonally polyestric. The breeding season in spring and summer and the winter anestrus are flanked by transitional periods. Endometrial diseases are a frequent cause of subfertility and have an economic impact on the horse breeding industry. They include different forms of endometrosis, endometritis, glandular maldifferentiation, and angiosis. Except for suppurative endometritis, these are subclinical and can only be diagnosed by the microscopic examination of an endometrial biopsy. Endometrosis is characterized by periglandular fibrosis and nonsuppurative endometritis by stromal infiltration with lymphocytes and plasma cells. The pathogenesis of endometrosis and nonsuppurative endometritis is still undetermined. Some mares are predisposed to persistent endometritis; this has likely a multifactorial etiology. Glandular differentiation has to be interpreted under consideration of the season. The presence of endometrial diseases is associated with alterations in the expression of several intra- and extracellular molecular markers. Some of them may have potential to be used as diagnostic biomarkers for equine endometrial health and disease. The aim of this review is to provide an overview on pathomorphological findings of equine endometrial diseases, to outline data on analyses of cellular and molecular mechanisms, and to discuss the impact of these data on reproduction and treatment.
Collapse
Affiliation(s)
- Sandra Schöniger
- Targos Molecular Pathology GmbH, Germaniastrasse 7, 34119 Kassel, Germany
- Correspondence:
| | - Heinz-Adolf Schoon
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 33, 04103 Leipzig, Germany;
| |
Collapse
|
42
|
Santiago CS, Albernaz ESS, Santos RTS, Guerra LHA, Santos FCA, Góes RM, Morielle-Versute E, Taboga SR, Beguelini MR. Evaluation of the uterine hormonal control of the bat Artibeus lituratus during the different phases of its reproductive cycle. J Morphol 2020; 281:302-315. [PMID: 31904879 DOI: 10.1002/jmor.21098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/03/2019] [Accepted: 12/22/2019] [Indexed: 12/24/2022]
Abstract
Artibeus lituratus is a frugivorous bat that directly assists in the restoration of degraded habitats through the effective dispersion of seeds and fruits. Given its great importance, this work aimed to evaluate the uterine hormonal control of A. lituratus during its different reproductive phases. The uteri of 30 sexually mature adult females, five specimens for each of the six sample groups (NON, nonreproductive; P1, initial pregnancy; P2, intermediate pregnancy; P3, advanced pregnancy; LAC, lactating; P + LAC, pregnant-lactating), were submitted to analyses of serum estradiol and progesterone concentrations, in addition to immunohistochemical analyses. Both estradiol and progesterone, gradually increased during pregnancy, with a marked significant increase in P3 females. Both returned to low levels in LAC-females; however, estradiol levels decreased further in P + LAC-females, while progesterone increased in the same group. In general, signs indicative of aromatase expression were observed in the endometrium of all analyzed groups and in the placenta of bats in the gestation groups. Similarly, ERα and PR were expressed in the myometrium, endometrium and placenta at varying levels of intensity. The results indicate that the uterine microenvironment of A. lituratus is directly regulated by serum concentrations of estradiol and progesterone, and fluctuations in these concentrations control morphological and physiological changes of this organ during different phases of the reproductive cycle. RESEARCH HIGHLIGHTS: Increases in serum concentrations of estradiol and progesterone coordinate the gestational period of A. lituratus. Estradiol activates ERα, stimulating cell proliferation in the uterus, in addition to activating the expression of PR, which trigger the quiescence of the myometrium and stimulation of the secretion and differentiation of the endometrium. Results showed several similarities to humans, indicating the use of A. lituratus as an animal model in reproductive studies.
Collapse
Affiliation(s)
- Cornélio S Santiago
- Center of Biological and Health Science, UFOB - Universidade Federal do Oeste da Bahia, Barreiras, Bahia, Brazil
| | - Edna S S Albernaz
- Center of Biological and Health Science, UFOB - Universidade Federal do Oeste da Bahia, Barreiras, Bahia, Brazil
| | - Renata T S Santos
- Center of Biological and Health Science, UFOB - Universidade Federal do Oeste da Bahia, Barreiras, Bahia, Brazil
| | - Luiz H A Guerra
- Department of Biology, UNESP - Univ. Estadual Paulista, São José do Rio Preto, São Paulo, Brazil
| | - Fernanda C A Santos
- Department of Histology and Embryology, UFG - Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Rejane M Góes
- Department of Biology, UNESP - Univ. Estadual Paulista, São José do Rio Preto, São Paulo, Brazil
| | - Eliana Morielle-Versute
- Department of Zoology and Botany, UNESP - Univ. Estadual Paulista, São José do Rio Preto, São Paulo, Brazil
| | - Sebastião R Taboga
- Department of Biology, UNESP - Univ. Estadual Paulista, São José do Rio Preto, São Paulo, Brazil
| | - Mateus R Beguelini
- Center of Biological and Health Science, UFOB - Universidade Federal do Oeste da Bahia, Barreiras, Bahia, Brazil
| |
Collapse
|
43
|
Tong Z, Liu Y, Yu X, Martinez JD, Xu J. The transcriptional co-activator NCOA6 promotes estrogen-induced GREB1 transcription by recruiting ERα and enhancing enhancer-promoter interactions. J Biol Chem 2019; 294:19667-19682. [PMID: 31744881 DOI: 10.1074/jbc.ra119.010704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/13/2019] [Indexed: 11/06/2022] Open
Abstract
Estrogen and its cognate receptor, ERα, regulate cell proliferation, differentiation, and carcinogenesis in the endometrium by controlling gene transcription. ERα requires co-activators to mediate transcription via mechanisms that are largely uncharacterized. Herein, using growth-regulating estrogen receptor binding 1 (GREB1) as an ERα target gene in Ishikawa cells, we demonstrate that nuclear receptor co-activator 6 (NCOA6) is essential for estradiol (E2)/ERα-activated GREB1 transcription. We found that NCOA6 associates with the GREB1 promoter and enhancer in an E2-independent manner and that NCOA6 knockout reduces chromatin looping, enhancer-promoter interactions, and basal GREB1 expression in the absence of E2. In the presence of E2, ERα bound the GREB1 enhancer and also associated with its promoter, and p300, myeloid/lymphoid or mixed-lineage leukemia protein 4 (MLL4), and RNA polymerase II were recruited to the GREB1 enhancer and promoter. Consequently, the levels of the histone modifications H3K4me1/3, H3K9ac, and H3K27ac were significantly increased; enhancer and promoter regions were transcribed; and GREB1 mRNA was robustly transcribed. NCOA6 knockout reduced ERα recruitment and abolished all of the aforementioned E2-induced events, making GREB1 completely insensitive to E2 induction. We also found that GREB1-deficient Ishikawa cells are much more resistant to chemotherapy and that human endometrial cancers with low GREB1 expression predict poor overall survival. These results indicate that NCOA6 has an essential role in ERα-mediated transcription by increasing enhancer-promoter interactions through chromatin looping and by recruiting RNA polymerase II and the histone-code modifiers p300 and MLL4. Moreover, GREB1 loss may predict chemoresistance of endometrial cancer.
Collapse
Affiliation(s)
- Zhangwei Tong
- Department of Molecular and Cellular Biology and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Yonghong Liu
- Department of Molecular and Cellular Biology and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Xiaobin Yu
- Department of Molecular and Cellular Biology and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Jarrod D Martinez
- Department of Molecular and Cellular Biology and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Jianming Xu
- Department of Molecular and Cellular Biology and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
44
|
Kelleher AM, DeMayo FJ, Spencer TE. Uterine Glands: Developmental Biology and Functional Roles in Pregnancy. Endocr Rev 2019; 40:1424-1445. [PMID: 31074826 PMCID: PMC6749889 DOI: 10.1210/er.2018-00281] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/15/2019] [Indexed: 12/18/2022]
Abstract
All mammalian uteri contain glands in the endometrium that develop only or primarily after birth. Gland development or adenogenesis in the postnatal uterus is intrinsically regulated by proliferation, cell-cell interactions, growth factors and their inhibitors, as well as transcription factors, including forkhead box A2 (FOXA2) and estrogen receptor α (ESR1). Extrinsic factors regulating adenogenesis originate from other organs, including the ovary, pituitary, and mammary gland. The infertility and recurrent pregnancy loss observed in uterine gland knockout sheep and mouse models support a primary role for secretions and products of the glands in pregnancy success. Recent studies in mice revealed that uterine glandular epithelia govern postimplantation pregnancy establishment through effects on stromal cell decidualization and placental development. In humans, uterine glands and, by inference, their secretions and products are hypothesized to be critical for blastocyst survival and implantation as well as embryo and placental development during the first trimester before the onset of fetal-maternal circulation. A variety of hormones and other factors from the ovary, placenta, and stromal cells impact secretory function of the uterine glands during pregnancy. This review summarizes new information related to the developmental biology of uterine glands and discusses novel perspectives on their functional roles in pregnancy establishment and success.
Collapse
Affiliation(s)
- Andrew M Kelleher
- Division of Animal Sciences, University of Missouri, Columbia, Missouri
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute on Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, Missouri.,Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, Missouri
| |
Collapse
|
45
|
Kim J, Cha S, Lee MY, Hwang YJ, Yang E, Choi D, Lee SH, Cheon YP. Chronic and Low Dose Exposure to Nonlyphenol or Di(2-Ethylhexyl) Phthalate Alters Cell Proliferation and the Localization of Steroid Hormone Receptors in Uterine Endometria in Mice. Dev Reprod 2019; 23:263-275. [PMID: 31660453 PMCID: PMC6812976 DOI: 10.12717/dr.2019.23.3.263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/30/2019] [Accepted: 09/19/2019] [Indexed: 02/04/2023]
Abstract
Based on our preliminary results, we examined the possible role of low-dose and
chronic-exposing of the chemicals those are known as endocrine disrupting
chemical (EDC), on the proliferation of uterine endometrium and the localization
of steroid receptors. Immunohistochemical or immunofluorochemical methodology
were employed to evaluate the localization of antigen identified by monoclonal
antibody Ki 67 protein (MKI67), estrogen receptor 1 (ESR1), estrogen receptor 2
(ESR2), and progesterone receptor (PGR). In 133 μg/L and 1,330
μg/L di(2-ethylhexyl) phthalate (DEHP) and 50 μg/L nonylphenol
(NP) groups, the ratio of MKI67 positive stromal cells was significantly
increased but not in 500 μg/L NP group. The ratios of MKI67 positive
glandular and luminal epithelial cells were also changed by the chronic
administration of NP and DEHP in tissue with dose specific manner. ESR1 signals
were localized in nucleus in glandular and luminal epithelia of control group
but its localization was mainly in cytoplasm in DEHP and NP administered groups.
On the other hand, it was decreased at nucleus of stromal cells in 1,330
μg/L DEHP group. The colocalization patterns of these nuclear receptors
were also modified by the administration of these chemicals. Such a tissue
specific and dose specific localization of ESR2 and PGR were detected as ESR1 in
all the uterine endometrial tissues. These results show that the chronic
lows-dose exposing of NP or DEHP modify the localization and colocalization of
ESRs and PGR, and of the proliferation patterns of the endometrial tissues.
Collapse
Affiliation(s)
- Juhye Kim
- Division of Developmental Biology and Physiology, Dept. of Biotechnology, Sungshin University, Seoul 02844, Korea
| | - Sunyeong Cha
- Division of Developmental Biology and Physiology, Dept. of Biotechnology, Sungshin University, Seoul 02844, Korea
| | - Min Young Lee
- Division of Developmental Biology and Physiology, Dept. of Biotechnology, Sungshin University, Seoul 02844, Korea
| | - Yeon Jeong Hwang
- Division of Developmental Biology and Physiology, Dept. of Biotechnology, Sungshin University, Seoul 02844, Korea
| | - Eunhyeok Yang
- Division of Developmental Biology and Physiology, Dept. of Biotechnology, Sungshin University, Seoul 02844, Korea
| | - Donchan Choi
- Dept. of Life Science, College of Environmental Sciences, Yong-In University, Yongin 17092, Korea
| | - Sung-Ho Lee
- Dept. of Biotechnology, Sangmyung University, Seoul 03016, Korea
| | - Yong-Pil Cheon
- Division of Developmental Biology and Physiology, Dept. of Biotechnology, Sungshin University, Seoul 02844, Korea
| |
Collapse
|
46
|
Marquardt RM, Kim TH, Shin JH, Jeong JW. Progesterone and Estrogen Signaling in the Endometrium: What Goes Wrong in Endometriosis? Int J Mol Sci 2019; 20:E3822. [PMID: 31387263 PMCID: PMC6695957 DOI: 10.3390/ijms20153822] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
In the healthy endometrium, progesterone and estrogen signaling coordinate in a tightly regulated, dynamic interplay to drive a normal menstrual cycle and promote an embryo-receptive state to allow implantation during the window of receptivity. It is well-established that progesterone and estrogen act primarily through their cognate receptors to set off cascades of signaling pathways and enact large-scale gene expression programs. In endometriosis, when endometrial tissue grows outside the uterine cavity, progesterone and estrogen signaling are disrupted, commonly resulting in progesterone resistance and estrogen dominance. This hormone imbalance leads to heightened inflammation and may also increase the pelvic pain of the disease and decrease endometrial receptivity to embryo implantation. This review focuses on the molecular mechanisms governing progesterone and estrogen signaling supporting endometrial function and how they become dysregulated in endometriosis. Understanding how these mechanisms contribute to the pelvic pain and infertility associated with endometriosis will open new avenues of targeted medical therapies to give relief to the millions of women suffering its effects.
Collapse
Affiliation(s)
- Ryan M Marquardt
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA
| | - Jung-Ho Shin
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Guro Hospital, Korea University Medical Center, Seoul 08318, Korea
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA.
| |
Collapse
|
47
|
Handgraaf S, Philippe J. The Role of Sexual Hormones on the Enteroinsular Axis. Endocr Rev 2019; 40:1152-1162. [PMID: 31074764 DOI: 10.1210/er.2019-00004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 05/03/2019] [Indexed: 12/17/2022]
Abstract
Sex steroid estrogens, androgens, and progesterone, produced by the gonads, which have long been considered as endocrine glands, are implicated in sexual differentiation, puberty, and reproduction. However, the impact of sex hormones goes beyond these effects through their role on energy metabolism. Indeed, sex hormones are important physiological regulators of glucose homeostasis and, in particular, of the enteroinsular axis. In this review, we describe the roles of estrogens, androgens, and progesterone on glucose homeostasis through their effects on pancreatic α- and β-cells, as well as on enteroendocrine L-cells, and their implications in hormonal biosynthesis and secretion. The analysis of their mechanisms of action with the dissection of the receptors implicated in the several protective effects could provide some new aspects of the fine-tuning of hormonal secretion under the influence of the sex. This knowledge paves the way to the understanding of transgender physiology and new potential therapeutics in the field of type 2 diabetes.
Collapse
Affiliation(s)
- Sandra Handgraaf
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension, and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland
| | - Jacques Philippe
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension, and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
48
|
Terakawa J, Serna VA, Taketo MM, Daikoku T, Suarez AA, Kurita T. Ovarian insufficiency and CTNNB1 mutations drive malignant transformation of endometrial hyperplasia with altered PTEN/PI3K activities. Proc Natl Acad Sci U S A 2019; 116:4528-4537. [PMID: 30782821 PMCID: PMC6410785 DOI: 10.1073/pnas.1814506116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Endometrioid endometrial carcinomas (EECs) carry multiple driver mutations even when they are low grade. However, the biological significance of these concurrent mutations is unknown. We explored the interactions among three signature EEC mutations: loss-of-function (LOF) mutations in PTEN, gain-of-function (GOF) mutations of phosphoinositide 3-kinase (PI3K), and CTNNB1 exon 3 mutations, utilizing in vivo mutagenesis of the mouse uterine epithelium. While epithelial cells with a monoallelic mutation in any one of three genes failed to propagate in the endometrium, any combination of two or more mutant alleles promoted the growth of epithelium, causing simple hyperplasia, in a dose-dependent manner. Notably, Ctnnb1 exon 3 deletion significantly increased the size of hyperplastic lesions by promoting the growth of PTEN LOF and/or PI3K GOF mutant cells through the activation of neoadenogenesis pathways. Although these three mutations were insufficient to cause EEC in intact female mice, castration triggered malignant transformation, leading to myometrial invasion and serosal metastasis. Treatment of castrated mice with progesterone or estradiol attenuated the neoplastic transformation. This study demonstrates that multiple driver mutations are required for premalignant cells to break the growth-repressing field effect of normal endometrium maintained by ovarian steroids and that CTNNB1 exon 3 mutations play critical roles in the growth of preneoplastic cells within the endometrium of premenopausal women and in the myometrial invasion of EECs in menopausal women.
Collapse
Affiliation(s)
- Jumpei Terakawa
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Vanida Ann Serna
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Makoto Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, 606-8506 Kyoto, Japan
| | - Takiko Daikoku
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, 920-8640 Kanazawa, Japan
| | - Adrian A Suarez
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
- Department of Pathology, Ohio State University, Columbus, OH 43210
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210;
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| |
Collapse
|
49
|
Shukla V, Kaushal JB, Sankhwar P, Manohar M, Dwivedi A. Inhibition of TPPP3 attenuates β-catenin/NF-κB/COX-2 signaling in endometrial stromal cells and impairs decidualization. J Endocrinol 2019; 240:417-429. [PMID: 30667362 DOI: 10.1530/joe-18-0459] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/03/2019] [Indexed: 12/13/2022]
Abstract
Embryo implantation and decidualization are critical events that occur during early pregnancy. Decidualization is synchronized by the crosstalk of progesterone and the cAMP signaling pathway. Previously, we confirmed the role of TPPP3 during embryo implantation in mice, but the underlying role and mechanism of TPPP3 in decidualization has not yet been understood. The current study was aimed to investigate the role of TPPP3 in decidualization in vivo and in vitro. For in vivo experiments, decidual reaction was artificially induced in the uteri of BALB/c mice. TPPP3 was found to be highly expressed during decidualization, whereas in the uteri receiving TPPP3 siRNA, decidualization was suppressed and the expression of β-catenin and decidual marker prolactin was reduced. In human endometrium, TPPP3 protein was found to be predominantly expressed in the mid-secretory phase (LH+7). In the primary culture of human endometrial stromal cells (hESCs), TPPP3 siRNA knockdown inhibited stromal-to-decidual cell transition and decreased the expression of the decidualization markers prolactin and IGFBP-1. Immunofluorescence and immunoblotting experiments revealed that TPPP3 siRNA knockdown suppressed the expression of β-catenin, NF-κB and COX-2 in hESCs during decidualization. TPPP3 inhibition also decreased NF-kB nuclear accumulation in hESCs and suppressed NF-κB transcriptional promoter activity. COX-2 expression was significantly decreased in the presence of a selective NF-kB inhibitor (QNZ) implicating that NF-kB is involved in COX-2 expression in hESCs undergoing decidualization. TUNEL assay and FACS analysis revealed that TPPP3 knockdown induced apoptosis and caused loss of mitochondrial membrane potential in hESCs. The study suggested that TPPP3 plays a significant role in decidualization and its inhibition leads to the suppression of β-catenin/NF-κB/COX-2 signaling along with the induction of mitochondria-dependent apoptosis.
Collapse
Affiliation(s)
- Vinay Shukla
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-CDRI Campus, Lucknow, India
| | - Jyoti Bala Kaushal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-CDRI Campus, Lucknow, India
| | - Pushplata Sankhwar
- Department of Obstetrics and Gynecology, King George's Medical University, Lucknow, India
| | - Murli Manohar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Anila Dwivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-CDRI Campus, Lucknow, India
| |
Collapse
|
50
|
Spencer TE, Kelleher AM, Bartol FF. Development and Function of Uterine Glands in Domestic Animals. Annu Rev Anim Biosci 2019; 7:125-147. [DOI: 10.1146/annurev-animal-020518-115321] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
All mammalian uteri contain glands that synthesize or transport and secrete substances into the uterine lumen. Uterine gland development, or adenogenesis, is uniquely a postnatal event in sheep and pigs and involves differentiation of glandular epithelium from luminal epithelium, followed by invagination and coiling morphogenesis throughout the stroma. Intrinsic transcription factors and extrinsic factors from the ovary and pituitary as well as the mammary gland (lactocrine) regulate uterine adenogenesis. Recurrent pregnancy loss is observed in the ovine uterine gland knockout sheep, providing unequivocal evidence that glands and their products are essential for fertility. Uterine gland hyperplasia and hypertrophy during pregnancy are controlled by sequential actions of hormones from the ovary and/or pituitary as well as the placenta. Gland-derived histotroph is transported by placental areolae for fetal growth. Increased knowledge of uterine gland biology is expected to improve pregnancy outcomes, as well as the health and productivity of mothers and their offspring.
Collapse
Affiliation(s)
- Thomas E. Spencer
- Division of Animal Sciences and Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri 65211, USA;,
| | - Andrew M. Kelleher
- Division of Animal Sciences and Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri 65211, USA;,
| | - Frank F. Bartol
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849-5517, USA
| |
Collapse
|