1
|
Srinivasan D, Raghavendhira H, Khan MZI, Kannan S, Bhaskaran RS. Cafeteria Diet-Induced Obesity Alters Uterine Function by Disrupting Insulin and Sex Steroid Actions in Rats. J Nutr 2025:S0022-3166(25)00181-6. [PMID: 40154886 DOI: 10.1016/j.tjnut.2025.03.029] [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: 10/23/2024] [Revised: 03/13/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND The cafeteria diet (CAFD) model leads to obesity in rats, disrupting glucose metabolism, hormonal balance, and ovarian function, which results in macrosomic offspring. Insulin and ovarian hormones are essential for uterine growth, but there is limited research on how CAFD-induced obesity affects uterine function by modulating hormonal concentrations. OBJECTIVE This study aimed to assess how CAFD-induced obesity impacts uterine function in adult female rats by analyzing concentration of ovarian steroids and insulin,along with uterine responses. METHODS Postweaning female Sprague-Dawley rats (22 d), were divided into a control group fed pelleted rat chow and an obese group fed energy-dense snacks (CAFD) and pelleted rat chow for 32 wk. Body weight, food intake, energy intake, and estrous cycles were monitored during the experiment. After the experimental period, the uterine tissues were evaluated histologically, and protein expression was analyzed using western blotting and immunofluorescence. Serum hormone concentrations were assessed by ELISA, and uterine oxidative stress markers (superoxide dismutase, catalase, reduced glutathione, lipid peroxidase, and vitamin C) were measured using spectrophotometric methods. RESULTS CAFD-fed rats exhibited increased body weight, BMI, and abdominal girth, along with hyperglycemia, extended estrous cycles averaging 8.9 d and 40% reduction in uterine weight (P < 0.0001). The expression of proliferating cell nuclear antigen was elevated (P < 0.0001), with a significant increase in uterine cell proliferation. Obese rats showed lower concentrations of superoxide dismutase, reduced glutathione, and vitamin C, whereas concentrations of lipid peroxidase and catalase were higher in uteri (P < 0.0001). Exposure to CAFD significantly reduced serum concentrations of prolactin, progesterone, and estradiol. Moreover, progesterone receptor and its target molecules (Indian hedgehog homolog, peroxisome proliferator-activated receptor γ, and prolactin receptor) were upregulated, whereas estrogen receptor-α (P < 0.0001) and its responsive molecules (vascular endothelial growth factor and homeobox A11) were downregulated (P < 0.0001). CONCLUSION Consumption of CAFD causes obesity, which reduces ovarian steroid and insulin secretion and increases oxidative stress in the rat uterus, altering the expression of key molecules vital for uterine function.
Collapse
Affiliation(s)
- Divya Srinivasan
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India.
| | - Harini Raghavendhira
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India.
| | - Mohammad Zafar Iqbal Khan
- Department of Anatomy, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India
| | - Saranya Kannan
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX, United States
| | - Ravi Sankar Bhaskaran
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India.
| |
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
|
Xie B, Chen Q, Dai Z, Jiang C, Chen X. Progesterone (P4) ameliorates cigarette smoke-induced chronic obstructive pulmonary disease (COPD). Mol Med 2024; 30:123. [PMID: 39138434 PMCID: PMC11323532 DOI: 10.1186/s10020-024-00883-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory lung disease associated with high morbidity and mortality worldwide. Oxidative injury and mitochondrial dysfunction in the airway epithelium are major events in COPD progression. METHODS AND RESULTS The therapeutic effects of Progesterone (P4) were investigated in vivo and in vitro in this study. In vivo, in a cigarette smoke (CS) exposure-induced COPD mouse model, P4 treatment significantly ameliorated CS exposure-induced physiological and pathological characteristics, including inflammatory cell infiltration and oxidative injury, in a dose-dependent manner. The c-MYC/SIRT1/PGC-1α pathway is involved in the protective function of P4 against CS-induced COPD. In vitro, P4 co-treatment significantly ameliorated H2O2-induced oxidative injury and mitochondrial dysfunctions by promoting cell proliferation, increasing mitochondrial membrane potential, decreasing ROS levels and apoptosis, and increasing ATP content. Moreover, P4 co-treatment partially attenuated H2O2-caused inhibition in Nrf1, Tfam, Mfn1, PGR-B, c-MYC, SIRT1, and PGC-1α levels. In BEAS-2B and ASM cells, the c-MYC/SIRT1 axis regulated P4's protective effects against H2O2-induced oxidative injury and mitochondrial dysfunctions. CONCLUSION P4 activates the c-MYC/SIRT1 axis, ameliorating CS-induced COPD and protecting both airway epithelial cells and smooth muscle cells against H2O2-induced oxidative damage. PGC-1α and downstream mitochondrial signaling pathways might be involved.
Collapse
Affiliation(s)
- Bin Xie
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Chen
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziyu Dai
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chen Jiang
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xi Chen
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
4
|
Roberson EC, Tran NK, Godambe AN, Mark H, Nguimtsop M, Rust T, Ung E, Barker LJ, Fitch RD, Wallingford JB. Hedgehog signaling is required for endometrial remodeling and myometrial homeostasis in the cycling mouse uterus. iScience 2023; 26:107993. [PMID: 37810243 PMCID: PMC10551904 DOI: 10.1016/j.isci.2023.107993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/24/2023] [Accepted: 09/16/2023] [Indexed: 10/10/2023] Open
Abstract
Decades of work demonstrate that the mammalian estrous cycle is controlled by cycling steroid hormones. However, the signaling mechanisms that act downstream, linking hormonal action to the physical remodeling of the cycling uterus, remain unclear. To address this issue, we analyzed gene expression at all stages of the mouse estrous cycle. Strikingly, we found that several genetic programs well-known to control tissue morphogenesis in developing embryos displayed cyclical patterns of expression. We find that most of the genetic architectures of Hedgehog signaling (ligands, receptors, effectors, and transcription factors) are transcribed cyclically in the uterus, and that conditional disruption of the Hedgehog receptor smoothened not only elicits a failure of normal cyclical thickening of the endometrial lining but also induces aberrant deformation of the uterine smooth muscle. Together, our data shed light on the mechanisms underlying normal uterine remodeling specifically and cyclical gene expression generally.
Collapse
Affiliation(s)
- Elle C. Roberson
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - Ngan Kim Tran
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Anushka N. Godambe
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Harrison Mark
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Michelle Nguimtsop
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Trinity Rust
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Elizabeth Ung
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - LeCaine J. Barker
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - Rebecca D. Fitch
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - John B. Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
5
|
Cheng J, Sha Z, Li J, Li B, Luo X, Zhang Z, Zhou Y, Chen S, Wang Y. Progress on the Role of Estrogen and Progesterone Signaling in Mouse Embryo Implantation and Decidualization. Reprod Sci 2023; 30:1746-1757. [PMID: 36694081 DOI: 10.1007/s43032-023-01169-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/28/2022] [Indexed: 01/25/2023]
Abstract
Embryo implantation and decidualization are key steps in establishing a successful pregnancy. Defects in embryo implantation and decidualization can cause a series of adverse chain reactions which can contribute to harmful pregnancy outcomes, such as embryo growth retardation, preeclampsia, miscarriage, premature birth, and so on. Approximately 75% of failed pregnancies are considered to be due to embryo implantation failure or defects. Decidualization, characterized by proliferation and differentiation of uterine stromal cells, is one of the essential conditions for blastocyst implantation, placental formation, and maintenance of pregnancy and is indispensable for the establishment of pregnancy in many species. Embryo implantation and decidualization are closely regulated by estrogen and progesterone secreted by the ovaries. Many cellular events and molecular signaling network pathways are involved in this process. This article reviews the recent advances in the molecular mechanisms of estrogen- and progesterone-regulating uterine receptivity establishment, blastocyst implantation, and decidualization, in order to better understand the underlying molecular mechanisms of hormonal regulation of embryo implantation and to develop new strategies for preventing or treating embryo implantation defects and improving the pregnancy rate of women.
Collapse
Affiliation(s)
- Jianghong Cheng
- Xi'An Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'An Medical University, Xi'An 710021, China
| | - Zizhuo Sha
- Xi'An Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'An Medical University, Xi'An 710021, China
| | - Junyang Li
- Xi'An Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'An Medical University, Xi'An 710021, China
| | - Bixuan Li
- Xi'An Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'An Medical University, Xi'An 710021, China
| | - Xianyang Luo
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Xiamen Key Laboratory of Otolaryngology Head and Neck Surgery, Xiamen, 361003, China.,Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, 350004, People's Republic of China
| | - Zhiming Zhang
- Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, 350004, People's Republic of China.,Department of Breast Surgery, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361003, People's Republic of China
| | - Yi Zhou
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Xiamen Key Laboratory of Otolaryngology Head and Neck Surgery, Xiamen, 361003, China.,Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, 350004, People's Republic of China
| | - Shuai Chen
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China. .,Xiamen Key Laboratory of Otolaryngology Head and Neck Surgery, Xiamen, 361003, China.
| | - Yang Wang
- Xi'An Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'An Medical University, Xi'An 710021, China.
| |
Collapse
|
6
|
Hwang YJ, Sung GJ, Marquardt R, Young SL, Lessey BA, Kim TH, Cheon YP, Jeong JW. SIRT1 plays an important role in implantation and decidualization during mouse early pregnancy. Biol Reprod 2022; 106:1072-1082. [PMID: 35134122 PMCID: PMC9198957 DOI: 10.1093/biolre/ioac026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/13/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Abstract
Sirtuin 1 (SIRT1) is a member of the sirtuin family that functions to deacetylate both histones and non-histone proteins. Previous studies have identified significant SIRT1 upregulation in eutopic endometrium from infertile women with endometriosis. However, SIRT1 function in the uterus has not been directly studied. Using immunochemistry analysis, we found SIRT1 to be most strongly expressed at GD4.5 and GD5.5 in decidualized cells and at GD7.5 in secondary decidual cells in mouse. To assess the role of SIRT1 in uterine function, we generated uterine Sirt1 conditional knockout mice (Pgrcre/+Sirt1f/f; Sirt1d/d). A 6-month fertility trial revealed that Sirt1d/d females were subfertile. Implantation site numbers were significantly decreased in Sirt1d/d mice compared with controls at GD5.5. Sirt1d/d implantation sites at GD4.5 could be divided into two groups, Group #1 with luminal closure and nonspecific COX2 expression compared with controls (14/20) and Group #2 with an open lumen and no COX2 (6/20). In Sirt1d/d Group #1, nuclear FOXO1 expression in luminal epithelial cells was significantly decreased. In Sirt1d/d Group #2, nuclear FOXO1 expression was almost completely absent, and there was strong PGR expression in epithelial cells. At GD5.5, stromal PGR and COX2 were significantly decreased in Sirt1d/d uterine in the areas surrounding the embryo compared with controls, indicating defective decidualization. An artificially induced decidualization test revealed that Sirt1d/d females showed defects in decidualization response. All together, these data suggest that SIRT1 is important for decidualization and contributes to preparing a receptive endometrium for successful implantation.
Collapse
Affiliation(s)
- Yeon Jeong Hwang
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
- Division of Developmental Biology and Physiology, Department of Biotechnology, Institute of Basic Sciences, Sungshin Women’s University, Seoul, South Korea
| | - Gi-Jun Sung
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Ryan Marquardt
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
- Cell and Molecular Biology Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Steven L Young
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of North Carolina, Chapel Hill, NC, USA
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertilithy, Atrium Health, Wake Forest Baptist, Winston-Salem, NC, USA
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Yong-Pil Cheon
- Division of Developmental Biology and Physiology, Department of Biotechnology, Institute of Basic Sciences, Sungshin Women’s University, Seoul, South Korea
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
7
|
Nuclear Receptors in Pregnancy and Outcomes: Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:3-19. [DOI: 10.1007/978-3-031-11836-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
8
|
Kim TH, Jeong JW. Proline-Rich Acidic Protein 1 (PRAP1) is a Target of ARID1A and PGR in the Murine Uterus. Dev Reprod 2019; 23:277-284. [PMID: 31660454 PMCID: PMC6812973 DOI: 10.12717/dr.2019.23.3.277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/13/2019] [Accepted: 09/05/2019] [Indexed: 12/05/2022]
Abstract
ARID1A and PGR plays an important role in embryo implantation and decidualization
during early pregnancy. Uterine specific Arid1a knockout
(Pgrcre/+Arid1af/f) mice
exhibit in non-receptive endometrium at day 3.5 of gestation (GD 3.5). In
previous studies, using transcriptomic analysis in the uterus of
Pgrcre/+Arid1af/f mice, we
identified proline-rich acidic protein 1 (PRAP1) as one of the
down-regulated genes by ARID1A in the uterus. In the present study, we performed
RT-qPCR and immunohistochemistry analysis to investigate the regulation of PRAP1
by ARID1A and determine expression patterns of PRAP1 in the uterus during early
pregnancy. During early pregnancy, PRAP1 expression was strong at day 0.5 of
gestation (GD 0.5) and then decreased at GD 3.5 in the epithelium and stroma.
After implantation, PRAP1 expression was remarkably reduced in the uterus.
However, the expression of PRAP1 at GD 3.5 was remarkably increased in the
Pgrcre/+Arid1a f/f mice.
To determine the ovarian steroid hormone regulation of PRAP1, we examined the
expression of PRAP1 in ovariectomized control,
Pgrcre/+Arid1af/f, and
progesterone receptor knock-out (PRKO) mice treated with progesterone. While
PRAP1 proteins were strongly expressed in the luminal and glandular epithelium
of control mice treated with vehicle, progesterone treatment suppressed the
expression of PRAP1. However, PRAP1 was not suppressed in both the
Pgrcre/+Arid1af/f and PRKO
mice compared to controls. Our results identified PRAP1 as a novel target of
ARID1A and PGR in the murine uterus.
Collapse
Affiliation(s)
- Tae Hoon Kim
- Dept. of Obstetrics, Gynecology & Reproductive Biology, Michigan State University College of Human Medicine, Grand Rapids, MI 49503, USA
| | - Jae-Wook Jeong
- Dept. of Obstetrics, Gynecology & Reproductive Biology, Michigan State University College of Human Medicine, Grand Rapids, MI 49503, USA
| |
Collapse
|
9
|
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: 129] [Impact Index Per Article: 21.5] [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
|
10
|
Zhang L, Feng Q, Wang Z, Liu P, Cui S. Progesterone receptor antagonist provides palliative effects for uterine leiomyoma through a Bcl-2/Beclin1-dependent mechanism. Biosci Rep 2019; 39:BSR20190094. [PMID: 31262976 PMCID: PMC6646233 DOI: 10.1042/bsr20190094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 12/22/2022] Open
Abstract
Uterine leiomyoma is the most common benign smooth muscle tumor of uterus in women of reproductive age, with a high lifetime incidence. Nowadays, the exploration on the pharmacotherapies, such as progesterone receptor antagonist (PRA) requires more attention. Hence, the current study aimed to examine whether mifepristone, a PRA, influences the autophagy and apoptosis of uterine leiomyoma cells. Primary uterine leiomyoma cells were collected from 36 patients diagnosed with uterine leiomyoma to establish PR-M-positive (PR-M[+]) cells. The lentiviral vector overexpressing or silencing PR-M was subsequently delivered into one part of PR-M(+) cells in order to evaluate the role of PR-M in PR-M(+) cells. The results obtained revealed that cell viability was increased, while cell autophagy and apoptosis were diminished in the PR-M(+) cells treated with overexpressed PR-M, whereby the Bcl-2 level was elevated and the level of Beclin1 was reduced. An opposite trends were identified following treatment with knockdown of PR-M. Mifepristone at different concentrations (low, moderate, or high) was then applied to treat another part of the PR-M(+) cells. Mifepristone was identified to promote cell autophagy and apoptosis, decrease Bcl-2 level and increase Beclin1 level, accompanied by weakened interaction between Bcl-2 and Beclin1. Moreover, these effects of mifepristone on PR-M(+) cells were enhanced with increasing of the concentration. Taken together, the present study present evidence indicates the ability of PRA to regulate the Bcl-2/Beclin1 axis, ultimately promoting the autophagy and apoptosis of uterine leiomyoma cells, highlighting that PRA serves as a promising therapeutic target for the treatment of uterine leiomyoma.
Collapse
Affiliation(s)
- Lindong Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Quanling Feng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhiting Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Pingping Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shihong Cui
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
11
|
Huang X, Zhong R, He X, Deng Q, Peng X, Li J, Luo X. Investigations on the mechanism of progesterone in inhibiting endometrial cancer cell cycle and viability via regulation of long noncoding RNA NEAT1/microRNA-146b-5p mediated Wnt/β-catenin signaling. IUBMB Life 2018; 71:223-234. [PMID: 30452118 DOI: 10.1002/iub.1959] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 11/12/2022]
Abstract
Progesterone is often used to protect the endometrium and prevent endometrial cancer. An intensive study on its molecular mechanism in endometrial cancer would contribute to the development of more promising therapies. Relevant lncRNAs and mRNAs expression data in endometrial cancer cell line Ishikawa pretreated and post-treated with progesterone were derived from Gene Expression Omnibus (accession no. GSE29435), and then we analyzed long noncoding RNAs and mRNAs with differential expressions in two different conditions. The Cytoscape software, TargetScan, miRanda, and Human microRNA Disease Database (HMDD) websites were employed. Gene set enrichment analysis (GSEA) was used to determine related Kyoto Encyclopedia of Genes and Genomes pathways alteration in Ishikawa cells treated with progesterone. In addition to bioinformatics analysis, Reverse Transcription-Polymerase Chain Reaction (RT-PCR), Western blot, and dual-luciferase reporter assays were performed. The impact of progesterone on cell propagation and cell cycle was testified by colony formation and flow cytometry analysis. LncRNA nuclear enriched abundant transcript 1 (NEAT1) was the most significantly downregulated lncRNA in endometrial cancer cells treated with progesterone. Lymphoid enhancing factor 1 (LEF1) was positively associated with NEAT1, and eventually hsa_miR-146b-5p was validated to target both LEF1 and NEAT1. Wnt/β-catenin signaling pathway was identified to involve in endometrial cancer. NEAT1 or LEF1 was overexpressed in endometrial cancer cells while downregulated following post-treatment with progesterone. Conversely, miR-146b-5p was notably decreased in Ishikawa cells while upregulated after treatment with progesterone. Downstream gene c-myc or MMP9 regulated by upstream gene LEF1 in Wnt/β-catenin signaling pathway was remarkably increased in Ishikawa cells and positively related with NEAT1. Progesterone inhibited cell cycle and viability through regulating NEAT1/miR-146b-5p axis via Wnt/β-catenin signaling pathway. Progesterone exerted suppressive influence on endometrial cancer progression via regulation of lncRNA NEAT1/miR-146b-5p-mediated Wnt/β-catenin signaling pathway, which might reveal new strategies for developing more effective therapeutics. © 2018 IUBMB Life, 71(1):223-234, 2019.
Collapse
Affiliation(s)
- Xiaohui Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People's Republic of China.,Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Rui Zhong
- Department of Ultrasound, Guangdong Province Traditional Chinese Medical Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Xiukui He
- Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Qingshan Deng
- Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Xiuhong Peng
- Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Jieming Li
- Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Xiping Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People's Republic of China.,Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
12
|
Ismail Madkour WA, Noah B, Abdel Hamid AMS, Zaheer H, Al-Bahr A, Shaeer M, Moawad A. Luteal phase support with estradiol and progesterone versus progesterone alone in GnRH antagonist ICSI cycles: a randomized controlled study. HUM FERTIL 2018; 19:142-9. [PMID: 27434094 DOI: 10.1080/14647273.2016.1200145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In vitro fertilization (IVF) cycles are associated with a defective luteal phase. Although progesterone supplementation to treat this problem is standard practice, estrogen addition is debatable. Our aim was to compare pregnancy outcomes in 220 patients undergoing antagonist intracytoplasmic sperm injection (ICSI) cycles protocol. The patients were randomly assigned into two equal groups to receive either vaginal progesterone alone (90 mg once daily) starting on the day of oocyte retrieval for up to 12 weeks if pregnancy occurred or estradiol addition (2 mg twice daily) starting on the same day and continuing up to seven weeks (foetal viability scan). Primary outcomes were pregnancy and ongoing pregnancy rates per embryo transfer. Secondary outcomes were implantation and early pregnancy loss rates. Pregnancy rates showed no significant difference between group 1 (39.09%) and 2 (43.63%) (p value = 0.3). Similarly, both groups were comparable regarding ongoing pregnancy rate (32.7% group 1 and 36.3% group 2, p value = 0.1). Implantation rates showed no difference between group 1 (19.25%) and group 2 (23.44%) (p value = 0.2). Early pregnancy loss rates were comparable, with 6.3% and 7.2% in groups 1 and 2, respectively, (p value = 0.4). In conclusion, the addition of 4 mg estrogen daily to progesterone for luteal support in antagonist ICSI cycles is not beneficial for pregnancy outcome.
Collapse
Affiliation(s)
- Wael A Ismail Madkour
- a Obstetrics and Gynecology Department , Ain-Shams University , Cairo , Egypt ;,b Dubai Gynecology and Fertility Centre , Dubai , UAE
| | - Bassel Noah
- b Dubai Gynecology and Fertility Centre , Dubai , UAE
| | - Amr M S Abdel Hamid
- a Obstetrics and Gynecology Department , Ain-Shams University , Cairo , Egypt ;,c IVF Unit , Enjab Hospital , Sharjah , UAE
| | - Hena Zaheer
- b Dubai Gynecology and Fertility Centre , Dubai , UAE
| | | | | | - Ashraf Moawad
- c IVF Unit , Enjab Hospital , Sharjah , UAE ;,d Obstetrics and Gynecology Department, Al-Azhar University , Cairo , Egypt
| |
Collapse
|
13
|
Le A, Wang ZH, Dai XY, Xiao TH, Zhuo R, Zhang B, Xiao Z, Fan X. Icaritin inhibits decidualization of endometrial stromal cells. Exp Ther Med 2017; 14:5949-5955. [PMID: 29285144 PMCID: PMC5740763 DOI: 10.3892/etm.2017.5278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 07/28/2017] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to investigate the effects of Icaritin on the proliferation and decidualization of endometrial stromal cells (ESCs). A total of 20 specimens of endometrium were collected during hysterectomy at the Gynecology Department of Shenzhen Nanshan People's Hospital (Shenzhen, China) between August 2014 and December 2015. The endometrium was digested with high concentrations of collagenase and DNase and filtered with meshes, and then the glandular epithelial and stromal cells were separated by the adhesion purification method. The purity of stromal cells was identified by vimetin and cytokeratin 7 immunostaining. The estradiol + progesterone (E2+P4) and/or cyclic adenosine monophosphate (cAMP) were added to induce an in vitro decidualization model, which was used to analyze the effect of Icaritin on the decidualization ability of the human ESCs. The decidualization markers of human ESCs, prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP-1), was analyzed by reverse-transcription quantitative polymerase chain reaction measurements of the mRNA levels, PRL immunostaining and ELISA analysis of the IGFBP-1 protein levels in the cells or cell culture supernatant separately. The results demonstrated that treatment with E2+P4 and/or cAMP for 96 h was able to induce decidualization in ESCs, and that the cells demonstrated polygon-shaped epithelioid changes. The cell nuclei revealed multinuclear changes, and the cells were also observed to be large and round in shape. The PRL expression and upregulated IGFBP-1 mRNA and protein levels in the E2+P4+cAMP treatment group indicated successful decidualization of the in vitro model. However, the addition of Icaritin inhibited the expression of PRL and IGFBP-1 mRNA, as well as IGFBP-1 protein in the induced ESCs compared with groups without Icaritin. These results suggest that Icaritin was able to inhibit the expression of decidualization-related genes in ESCs in vitro. However, the exact mechanisms require further investigation.
Collapse
Affiliation(s)
- Aiwen Le
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Nanshan People's Hospital of Guangdong Medical University, Shenzhen, Guangdong 518052, P.R. China
| | - Zhong Hai Wang
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Nanshan People's Hospital of Guangdong Medical University, Shenzhen, Guangdong 518052, P.R. China
| | - Xiao Yun Dai
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Nanshan People's Hospital of Guangdong Medical University, Shenzhen, Guangdong 518052, P.R. China
| | - Tian Hui Xiao
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Nanshan People's Hospital of Guangdong Medical University, Shenzhen, Guangdong 518052, P.R. China
| | - Rong Zhuo
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Nanshan People's Hospital of Guangdong Medical University, Shenzhen, Guangdong 518052, P.R. China
| | - Baozhen Zhang
- Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
| | - Zhonglin Xiao
- Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
| | - Xiujun Fan
- Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
| |
Collapse
|
14
|
Kang X, Zhang X, Zhao A. Macrophage depletion and TNF-α inhibition prevent resorption in CBA/J × DBA/2 model of CpG-induced abortion. Biochem Biophys Res Commun 2015; 469:704-10. [PMID: 26686416 DOI: 10.1016/j.bbrc.2015.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 12/04/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To elucidate the mechanism by which embryo-resorption was enhanced by pathogenic CpG ODN motif in abortion-prone CBA/J × DBA/2 model and to develop a counter strategy for normal pregnancy outcome. METHODS This is an animal model-based study. Abortion-prone model is established by CBA/J × DBA/2. An infection was mimicked by CpG ODN injection. RESULTS Embryo-resorption was readily induced by CpG ODN in low doses of CpG ODN (∼25 μg/dam) when intraperitoneally (IP) injected on gestational day(gd) 6.5 in male DBA/2 mated CBA/J female mice. A more modest decline in Progesterone(P4), but not Estrogen(E2) was observed after exposure to CpG ODN in the model. P4 supplement fail to improve pregnancy outcomes, even at pharmocology dose. CpG ODN-induced fetal resorption is prevented by the treatment of anti-F4/80 or by that of anti-TNFα.In the implantation sites, the treatment of anti-F4/80 inhibits the increase both of F4/80(+) macrophage proportion and TNF-αexpression level which are induced by CpG ODN. The anti-TNFαtreatment also recovers CpG ODN-induced reduction of CD4(+)Foxp3(+) T cells. CONCLUSION Circulating P4 is not responsible for the process by which CpG ODN-induced embryonic resorption in an abortion-prone mice. Macrophage depletion and TNF-α inhibition are really noteworthy for CpG ODN-induced pregnancy disruption.
Collapse
Affiliation(s)
- Xiaomin Kang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, PR China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, PR China
| | - Xiaoxin Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, PR China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, PR China
| | - Aimin Zhao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, PR China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, PR China.
| |
Collapse
|
15
|
Villanueva H, Visbal AP, Obeid NF, Ta AQ, Faruki AA, Wu MF, Hilsenbeck SG, Shaw CA, Yu P, Plummer NW, Birnbaumer L, Lewis MT. An essential role for Gα(i2) in Smoothened-stimulated epithelial cell proliferation in the mammary gland. Sci Signal 2015; 8:ra92. [PMID: 26373672 DOI: 10.1126/scisignal.aaa7355] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hedgehog (Hh) signaling is critical for organogenesis, tissue homeostasis, and stem cell maintenance. The gene encoding Smoothened (SMO), the primary effector of Hh signaling, is expressed aberrantly in human breast cancer, as well as in other cancers. In mice that express a constitutively active form of SMO that does not require Hh stimulation in mammary glands, the cells near the transgenic cells proliferate and participate in hyperplasia formation. Although SMO is a seven-transmembrane receptor like G protein-coupled receptors (GPCRs), SMO-mediated activation of the Gli family of transcription factors is not known to involve G proteins. However, data from Drosophila and mammalian cell lines indicate that SMO functions as a GPCR that couples to heterotrimeric G proteins of the pertussis toxin (PTX)-sensitive Gαi class. Using genetically modified mice, we demonstrated that SMO signaling through G proteins occurred in the mammary gland in vivo. SMO-induced stimulation of proliferation was PTX-sensitive and required Gαi2, but not Gαi1, Gαi3, or activation of Gli1 or Gli2. Our findings show that activated SMO functions as a GPCR to stimulate proliferation in vivo, a finding that may have clinical importance because most SMO-targeted agents have been selected based largely on their ability to block Gli-mediated transcription.
Collapse
Affiliation(s)
- Hugo Villanueva
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA. Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Adriana P Visbal
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA. Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nadine F Obeid
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrew Q Ta
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Adeel A Faruki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng-Fen Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA. Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chad A Shaw
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peng Yu
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Nicholas W Plummer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Michael T Lewis
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA. Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA. Department of Radiology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
16
|
Moravek MB, Yin P, Ono M, Coon JS, Dyson MT, Navarro A, Marsh EE, Chakravarti D, Kim JJ, Wei JJ, Bulun SE. Ovarian steroids, stem cells and uterine leiomyoma: therapeutic implications. Hum Reprod Update 2014; 21:1-12. [PMID: 25205766 DOI: 10.1093/humupd/dmu048] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Uterine leiomyoma is the most common benign tumor in women and is thought to arise from the clonal expansion of a single myometrial smooth muscle cell transformed by a cellular insult. Leiomyomas cause a variety of symptoms, including abnormal uterine bleeding, pelvic pain, bladder or bowel dysfunction, and recurrent pregnancy loss, and are the most common indication for hysterectomy in the USA. A slow rate of cell proliferation, combined with the production of copious amounts of extracellular matrix, accounts for tumor expansion. A common salient feature of leiomyomas is their responsiveness to steroid hormones, thus providing an opportunity for intervention. METHODS A comprehensive search of PUBMED was conducted to identify peer-reviewed literature published since 1980 pertinent to the roles of steroid hormones and somatic stem cells in leiomyoma, including literature on therapeutics that target steroid hormone action in leiomyoma. Reviewed articles were restricted to English language only. Studies in both animals and humans were reviewed for the manuscript. RESULTS Estrogen stimulates the growth of leiomyomas, which are exposed to this hormone not only through ovarian steroidogenesis, but also through local conversion of androgens by aromatase within the tumors themselves. The primary action of estrogen, together with its receptor estrogen receptor α (ERα), is likely mediated via induction of progesterone receptor (PR) expression, thereby allowing leiomyoma responsiveness to progesterone. Progesterone has been shown to stimulate the growth of leiomyoma through a set of key genes that regulate both apoptosis and proliferation. Given these findings, aromatase inhibitors and antiprogestins have been developed for the treatment of leiomyoma, but neither treatment results in complete regression of leiomyoma, and tumors recur after treatment is stopped. Recently, distinct cell populations were discovered in leiomyomas; a small population showed stem-progenitor cell properties, and was found to be essential for ovarian steroid-dependent growth of leiomyomas. Interestingly, these stem-progenitor cells were deficient in ERα and PR and instead relied on the strikingly higher levels of these receptors in surrounding differentiated cells to mediate estrogen and progesterone action via paracrine signaling. CONCLUSIONS It has been well established that estrogen and progesterone are involved in the proliferation and maintenance of uterine leiomyoma, and the majority of medical treatments currently available for leiomyoma work by inhibiting steroid hormone production or action. A pitfall of these therapeutics is that they decrease leiomyoma size, but do not completely eradicate them, and tumors tend to regrow once treatment is stopped. The recent discovery of stem cells and their paracrine interactions with more differentiated cell populations within leiomyoma has the potential to provide the missing link between developing therapeutics that temper leiomyoma growth and those that eradicate them.
Collapse
Affiliation(s)
- Molly B Moravek
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Ping Yin
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - John S Coon
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Matthew T Dyson
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Antonia Navarro
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Erica E Marsh
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Debabrata Chakravarti
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - J Julie Kim
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Jian-Jun Wei
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA Department of Pathology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Serdar E Bulun
- Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
17
|
Knoedler JR, Denver RJ. Krüppel-like factors are effectors of nuclear receptor signaling. Gen Comp Endocrinol 2014; 203:49-59. [PMID: 24642391 PMCID: PMC4339045 DOI: 10.1016/j.ygcen.2014.03.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 02/28/2014] [Accepted: 03/02/2014] [Indexed: 01/09/2023]
Abstract
Binding of steroid and thyroid hormones to their cognate nuclear receptors (NRs) impacts virtually every aspect of postembryonic development, physiology and behavior, and inappropriate signaling by NRs may contribute to disease. While NRs regulate genes by direct binding to hormone response elements in the genome, their actions may depend on the activity of other transcription factors (TFs) that may or may not bind DNA. The Krüppel-like family of transcription factors (KLF) is an evolutionarily conserved class of DNA-binding proteins that influence many aspects of development and physiology. Several members of this family have been shown to play diverse roles in NR signaling. For example, KLFs (1) act as accessory transcription factors for NR actions, (2) regulate expression of NR genes, and (3) as gene products of primary NR response genes function as key players in NR-dependent transcriptional networks. In mouse models, deletion of different KLFs leads to aberrant transcriptional and physiological responses to hormones, underscoring the importance of these proteins in the regulation of hormonal signaling. Understanding the functional relationships between NRs and KLFs will yield important insights into mechanisms of NR signaling. In this review we present a conceptual framework for understanding how KLFs participate in NR signaling, and we provide examples of how these proteins function to effect hormone action.
Collapse
Affiliation(s)
- Joseph R Knoedler
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Robert J Denver
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109-1048, USA; Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA.
| |
Collapse
|
18
|
Zaitseva M, Holdsworth-Carson SJ, Waldrip L, Nevzorova J, Martelotto L, Vollenhoven BJ, Rogers PAW. Aberrant expression and regulation of NR2F2 and CTNNB1 in uterine fibroids. Reproduction 2013; 146:91-102. [DOI: 10.1530/rep-13-0087] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Uterine fibroids are the most common benign tumour afflicting women of reproductive age. Despite the large healthcare burden caused by fibroids, there is only limited understanding of the molecular mechanisms that drive fibroid pathophysiology. Although a large number of genes are differentially expressed in fibroids compared with myometrium, it is likely that most of these differences are a consequence of the fibroid presence and are not causal. The aim of this study was to investigate the expression and regulation of NR2F2 and CTNNB1 based on their potential causal role in uterine fibroid pathophysiology. We used real-time quantitative RT-PCR, western blotting and immunohistochemistry to describe the expression of NR2F2 and CTNNB1 in matched human uterine fibroid and myometrial tissues. Primary myometrial and fibroid smooth muscle cell cultures were treated with progesterone and/or retinoic acid (RA) and sonic hedgehog (SHH) conditioned media to investigate regulatory pathways for these proteins. We showed that NR2F2 and CTNNB1 are aberrantly expressed in fibroid tissue compared with matched myometrium, with strong blood vessel-specific localisation. Although the SHH pathway was shown to be active in myometrial and fibroid primary cultures, it did not regulateNR2F2orCTNNB1mRNA expression. However, progesterone and RA combined regulatedNR2F2mRNA, but notCTNNB1, in myometrial but not fibroid primary cultures. In conclusion, we demonstrate aberrant expression and regulation of NR2F2 and CTNNB1 in uterine fibroids compared with normal myometrium, consistent with the hypothesis that these factors may play a causal role uterine fibroid development.
Collapse
|
19
|
Kim JJ, Kurita T, Bulun SE. Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocr Rev 2013; 34:130-62. [PMID: 23303565 PMCID: PMC3565104 DOI: 10.1210/er.2012-1043] [Citation(s) in RCA: 350] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 09/17/2012] [Indexed: 12/19/2022]
Abstract
Progesterone receptor (PR) mediates the actions of the ovarian steroid progesterone, which together with estradiol regulates gonadotropin secretion, prepares the endometrium for implantation, maintains pregnancy, and differentiates breast tissue. Separation of estrogen and progesterone actions in hormone-responsive tissues remains a challenge. Pathologies of the uterus and breast, including endometrial cancer, endometriosis, uterine fibroids, and breast cancer, are highly associated with estrogen, considered to be the mitogenic factor. Emerging evidence supports distinct roles of progesterone and its influence on the pathogenesis of these diseases. Progesterone antagonizes estrogen-driven growth in the endometrium, and insufficient progesterone action strikingly increases the risk of endometrial cancer. In endometriosis, eutopic and ectopic tissues do not respond sufficiently to progesterone and are considered to be progesterone-resistant, which contributes to proliferation and survival. In uterine fibroids, progesterone promotes growth by increasing proliferation, cellular hypertrophy, and deposition of extracellular matrix. In normal mammary tissue and breast cancer, progesterone is pro-proliferative and carcinogenic. A key difference between these tissues that could explain the diverse effects of progesterone is the paracrine interactions of PR-expressing stroma and epithelium. Normal endometrium is a mucosa containing large quantities of distinct stromal cells with abundant PR, which influences epithelial cell proliferation and differentiation and protects against carcinogenic transformation. In contrast, the primary target cells of progesterone in the breast and fibroids are the mammary epithelial cells and the leiomyoma cells, which lack specifically organized stromal components with significant PR expression. This review provides a unifying perspective for the diverse effects of progesterone across human tissues and diseases.
Collapse
Affiliation(s)
- J Julie Kim
- Division of Reproductive Biology Research, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | |
Collapse
|
20
|
Large MJ, DeMayo FJ. The regulation of embryo implantation and endometrial decidualization by progesterone receptor signaling. Mol Cell Endocrinol 2012; 358:155-65. [PMID: 21821095 PMCID: PMC3256265 DOI: 10.1016/j.mce.2011.07.027] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 07/09/2011] [Accepted: 07/12/2011] [Indexed: 12/17/2022]
Abstract
During the early stages of pregnancy, fertilized embryos must attach to the uterine epithelium, invade into the underlying uterine stroma, and the stroma must then differentiate in a process termed decidualization in order for a successful pregnancy to be initiated. The steroid hormone progesterone (P4) is an integral mediator of these early pregnancy events, exerting its effects via the progesterone receptor (PR). Insights gained from the use of mouse models and genomic profiling has identified many of the key molecules enlisted by PR to execute the paradigm of early pregnancy. This review describes several of the molecules through which the PR exerts its pleiotropic effects including ligands, receptors, chaperones, signaling proteins and transcription factors. Understanding these molecules and their concatenation is of vital importance to our ability to clinically treat reproductive health problems like infertility and endometriosis.
Collapse
Affiliation(s)
- Michael J. Large
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030 (USA)
| | - Francesco J. DeMayo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030 (USA)
| |
Collapse
|
21
|
Lipopolysaccharide-induced modulation in the expression of progesterone receptor and estradiol receptor leads to early pregnancy loss in mouse. ZYGOTE 2012; 21:337-44. [DOI: 10.1017/s0967199412000330] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SummaryThe objective of the present study was to investigate the effect of Gram-negative bacteria infection on ovarian steroid receptors, i.e. progesterone receptor (PR) and estradiol receptor (ER) during preimplantation days of pregnancy. A well established mouse model of Gram-negative bacteria infection was used to test this objective. Mice were treated with normal saline or lipopolysaccharide (LPS) on day 0.5 of pregnancy and used to collect embryos and uterine horns on day 1.5 to day 4.42 preimplantation day of pregnancy. Total RNA was extracted and reverse-transcription polymerase chain reaction (PCR) was performed to check the expression of PR and ER genes. The mRNA expression of PR and ER was altered in embryos and uterus of LPS-treated animals during preimplantation days of pregnancy studied. These results suggest that PR and ER play an important role in Gram-negative bacteria infection and induced implantation failure in mouse.
Collapse
|
22
|
Hu Q, Yu L, Chen R, Wang YL, Ji L, Zhang Y, Xie Y, Liao QP. 5-aza-2'-deoxycytidine improves the sensitivity of endometrial cancer cells to progesterone therapy. Int J Gynecol Cancer 2012; 22:951-9. [PMID: 22683940 DOI: 10.1097/igc.0b013e3182540160] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Progesterone has been proven to have limited effects on endometrial cancers (ECs), mainly owing to the down-regulation of progesterone receptor (PR). Here, we explored whether 5-aza-2'-deoxycytidine (5-aza-CdR), a demethylating agent, could enhance the susceptibility of EC cells to medroxyprogesterone acetate (MPA). METHODS Ishikawa and KLE cell lines were treated with 5-aza-CdR and/or MPA. The expression of PR, PR target genes, and matrix metalloproteinase (MMP) was investigated by real-time polymerase chain reaction and Western blot. Promoter methylation was detected by methylation-specific polymerase chain reaction. The effects of 5-aza-CdR and/or MPA on cell proliferation, apoptosis, and invasion of EC cells were evaluated by 2-(4-Iodophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium assay, flow cytometry, invasion assay, and gelatin zymography, respectively. RESULTS 5-Aza-2'-deoxycytidine significantly increased the expression of PR and its downstream targets by demethylating PR promoter in both Ishikawa and KLE cells. 5-Aza-2'-deoxycytidine combined with MPA synergistically suppressed the EC cell growth by inducing cell cycle arrest at G2/M phase and apoptosis. Furthermore, 5-aza-CdR synergized with MPA to inhibit the invasion of EC cells, perhaps owing to the down-regulation of MMP-2 and MMP-9 expression and activity. CONCLUSIONS 5-Aza-2'-deoxycytidine and MPA synergistically inhibit EC cell growth and invasion. Their combined use may provide a new effective therapeutic opportunity for endometrial carcinoma.
Collapse
Affiliation(s)
- Qian Hu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
KLF15 negatively regulates estrogen-induced epithelial cell proliferation by inhibition of DNA replication licensing. Proc Natl Acad Sci U S A 2012; 109:E1334-43. [PMID: 22538816 DOI: 10.1073/pnas.1118515109] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In the epithelial compartment of the uterus, estradiol-17β (E(2)) induces cell proliferation while progesterone (P(4)) inhibits this response and causes differentiation of the cells. In this study, we identified the mechanism whereby E(2) and P(4) reciprocally regulate the expression of minichromosome maintenance (MCM)-2, a protein that is an essential component of the hexameric MCM-2 to 7 complex required for DNA synthesis initiation. We show in the uterine epithelium that Kruppel-like transcription (KLF) factors, KLF 4 and 15, are inversely expressed; most importantly, they bind to the Mcm2 promoter under the regulation of E(2) and P(4)E(2), respectively. After P(4)E(2) exposure and in contrast to E(2) treated mice, the Mcm2 promoter displays increased histone 3 (H3) methylation and the recruitment of histone deacetylase 1 and 3 with the concomitant deacetylation of H3. This increased methylation and decreased acetylation is associated with an inhibition of RNA polymerase II binding, indicating an inactive Mcm2 promoter following P(4)E(2) treatment. Using transient transfection assays in the Ishikawa endometrial cell line, we demonstrate that Mcm2 promoter activity is hormonally stimulated by E(2) and that KLF15 inhibits this E(2) enhanced transcription. KLF15 expression also blocks Ishikawa cell proliferation through inhibition of MCM2 protein level. Importantly, in vivo expression of KLF15 in an estrogenized uterus mimics P(4)'s action by inhibiting E(2)-induced uterine epithelial MCM-2 expression and DNA synthesis. KLF15 is therefore a downstream physiological mediator of progesterone's cell cycle inhibitory action in the uterine epithelium.
Collapse
|
24
|
Migone FF, Ren Y, Cowan RG, Harman RM, Nikitin AY, Quirk SM. Dominant activation of the hedgehog signaling pathway alters development of the female reproductive tract. Genesis 2011; 50:28-40. [PMID: 21809434 DOI: 10.1002/dvg.20786] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 06/08/2011] [Accepted: 07/01/2011] [Indexed: 12/16/2022]
Abstract
The role of hedgehog (HH) signaling in reproductive tract development was studied in mice in which a dominant active allele of the signal transducer smoothened (SmoM2) was conditionally expressed in the Müllerian duct and ovary. Mutant females are infertile, primarily because they fail to ovulate. Levels of mRNA for targets of HH signaling, Gli1, Ptch1, and Hhip, were elevated in reproductive tracts of 24-day-old mutant mice, confirming overactivation of HH signaling. The tracts of mutant mice developed abnormally. The uterine luminal epithelium had a simple columnar morphology in control mice, but in mutants contained stratified squamous cells typical of the cervix and vagina. In mutant mice, the number of uterine glands were reduced and the oviducts were not coiled. Expression of genes within the Hox and Wnt families that regulate patterning of the reproductive tract were altered. Hoxa13, which is normally expressed primarily in the vagina and cervix, was expressed at 12-fold higher levels in the uterus of mutant mice compared with controls. Wnt5a, which is required for development of the cervix and vagina and postnatal differentiation of the uterus, was expressed at higher levels in the oviduct and uterus of mutant mice compared with controls. Mating mutant females with fertile or vasectomized males induced a severe inflammatory response in the tract. In summary, overactivation of HH signaling causes aberrant development of the reproductive tract. The phenotype observed could be mediated by ectopic expression of Hoxa13 in the uterus and elevated levels of Wnt5a in the oviducts and uterus.
Collapse
Affiliation(s)
- Fernando F Migone
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York
| | | | | | | | | | | |
Collapse
|
25
|
Afshar Y, Jeong JW, Roqueiro D, DeMayo F, Lydon J, Radtke F, Radnor R, Miele L, Fazleabas A. Notch1 mediates uterine stromal differentiation and is critical for complete decidualization in the mouse. FASEB J 2011; 26:282-94. [PMID: 21990372 DOI: 10.1096/fj.11-184663] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Uterine receptivity implies a dialogue between the hormonally primed maternal endometrium and the free-floating blastocyst. Endometrial stromal cells proliferate, avert apoptosis, and undergo decidualization in preparation for implantation; however, the molecular mechanisms that underlie differentiation into the decidual phenotype remain largely undefined. The Notch family of transmembrane receptors transduce extracellular signals responsible for cell survival, cell-to-cell communication, and differentiation, all fundamental processes for decidualization and pregnancy. Using a murine artificial decidualization model, pharmacological inhibition of Notch signaling by γ-secretase inhibition resulted in a significantly decreased deciduoma. Furthermore, a progesterone receptor (PR)-Cre Notch1 bigenic (Notch1(d/d)) confirmed a Notch1-dependent hypomorphic decidual phenotype. Microarray and pathway analysis, following Notch1 ablation, demonstrated significantly altered signaling repertoire. Concomitantly, hierarchical clustering demonstrated Notch1-dependent differences in gene expression. Uteri deprived of Notch1 signaling demonstrated decreased cellular proliferation; namely, reduced proliferation-specific antigen, Ki67, altered p21, cdk6, and cyclinD activity and an increased apoptotic-profile, cleaved caspase-3, Bad, and attenuated Bcl2. The results demonstrate that the preimplantation uterus relies on Notch signaling to inhibit apoptosis of stromal fibroblasts and regulate cell cycle progression, which together promotes successful decidualization. In summary, Notch1 signaling modulates multiple signaling mechanisms crucial for decidualization and these studies provide additional perspectives to the coordination of multiple signaling modalities required during decidualization.
Collapse
Affiliation(s)
- Yalda Afshar
- Department of Physiology and Biophysics, University of Illinois, Chicago, IL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lipopolysaccharide induces alterations in ovaries and serum level of progesterone and 17β-estradiol in the mouse. Fertil Steril 2011; 95:1471-4. [DOI: 10.1016/j.fertnstert.2010.08.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 07/29/2010] [Accepted: 08/19/2010] [Indexed: 12/29/2022]
|
27
|
Konno T, Graham AR, Rempel LA, Ho-Chen JK, Alam SMK, Bu P, Rumi MAK, Soares MJ. Subfertility linked to combined luteal insufficiency and uterine progesterone resistance. Endocrinology 2010; 151:4537-50. [PMID: 20660062 PMCID: PMC2940508 DOI: 10.1210/en.2010-0440] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Early pregnancy loss is common and can be caused by a range of factors. The Brown Norway (BN) rat exhibits reproductive dysfunction characterized by small litter size and pregnancy failure and represents a model for investigating early pregnancy loss. In this study, we investigated the establishment of pregnancy in the BN rat and gained insight into mechanisms causing its subfertility. Early stages of BN uteroplacental organization are unique. The BN primordial placenta is restricted in its development and correlates with limited BN uterine decidual development. BN uterine decidua was shown to be both structurally and functionally distinct and correlated with decreased circulating progesterone (P4) levels. Ovarian anomalies were also apparent in BN rats and included decreased ovulation rates and decreased transcript levels for some steroidogenic enzymes. Attempts to rescue the BN uterine decidual phenotype with steroid hormone therapy were ineffective. BN uteri were shown to exhibit reduced responsiveness to P4 but not to 17beta-estradiol. P4 resistance was associated with decreased transcript levels for the P4 receptor (Pgr), a P4 receptor chaperone (Fkbp4), and P4 receptor coactivators (Ncoa1 and Ncoa2). In summary, the BN rat exhibits luteal insufficiency and uterine P4 resistance, which profoundly affects its ability to reproduce.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Line, Tumor
- Cells, Cultured
- Corpus Luteum/drug effects
- Corpus Luteum/metabolism
- Decidua/metabolism
- Estradiol/pharmacology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Humans
- Infertility/genetics
- Infertility/metabolism
- Luciferases/genetics
- Luciferases/metabolism
- Male
- Progesterone/blood
- Progesterone/metabolism
- Progesterone/pharmacology
- Promoter Regions, Genetic/genetics
- Rats
- Rats, Inbred BN
- Rats, Inbred Dahl
- Rats, Inbred F344
- Rats, Sprague-Dawley
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Sequence Analysis, DNA
- Stromal Cells/cytology
- Stromal Cells/metabolism
- Uterus/cytology
- Uterus/drug effects
- Uterus/metabolism
Collapse
Affiliation(s)
- Toshihiro Konno
- Institute of Maternal-Fetal Biology, and Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Franco HL, Lee KY, Broaddus RR, White LD, Lanske B, Lydon JP, Jeong JW, DeMayo FJ. Ablation of Indian hedgehog in the murine uterus results in decreased cell cycle progression, aberrant epidermal growth factor signaling, and increased estrogen signaling. Biol Reprod 2010; 82:783-90. [PMID: 20056671 PMCID: PMC2842491 DOI: 10.1095/biolreprod.109.080259] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Conditional ablation of Indian hedgehog (Ihh) in the murine uterus results in mice that are sterile because of defects in embryo implantation. We performed microarray analysis on these mice at the time point at which the Ihh target genes are induced by the administration of exogenous hormone to mimic Day 3.5 of pregnancy. This analysis identified 863 genes altered by the conditional ablation of Ihh. Of these, genes that regulated the cell cycle were overrepresented. In addition, genes involved in epidermal growth factor (EGF) and estrogen (E2) signaling were found to be deregulated upon Ihh ablation. Furthermore, upon conditional ablation of Ihh, 15-mo-old mice exhibited hallmarks of estrogenized uteri, such as cystically dilated glands and hyalinized stroma. Thus, Ihh regulates embryo implantation by having an impact on the cell cycle, EGF signaling, and E2 signaling.
Collapse
Affiliation(s)
- Heather L. Franco
- Departments of Molecular and Cellular Biology and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Kevin Y. Lee
- Departments of Molecular and Cellular Biology and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Russell R. Broaddus
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lisa D. White
- Departments of Molecular and Cellular Biology and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Beate Lanske
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - John P. Lydon
- Departments of Molecular and Cellular Biology and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Jae-Wook Jeong
- Departments of Molecular and Cellular Biology and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Francesco J. DeMayo
- Departments of Molecular and Cellular Biology and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas,Correspondence: Francesco J. DeMayo, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030. FAX: 713 790 1275; e-mail:
| |
Collapse
|
29
|
Varricchio E, Coccia E, Putti R, Paolucci M. The olfactory organ of the trout Salmo trutta fario: a novel localization for a progestin receptor. Microsc Res Tech 2010; 73:206-14. [PMID: 19725068 DOI: 10.1002/jemt.20776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A progestin receptor (PR) has been detected in the olfactory organ of the trout Salmo trutta fario. The specificity of this receptor was high for 17alpha,20beta-dihydroxy-4-pregnen-3-one (17alpha,20beta-DP), but it also bound 17alpha-hydroxy-progesterone (17alpha-OHP) and 21-hydroxyprogesterone (21-OHP), even when present at low concentrations (10-fold in relative binding affinity assay). Progesterone (P) competed effectively at much higher concentrations (1,000-fold in relative binding affinity assay). Immunohistochemical studies carried out with three different monoclonal antibodies against human progesterone receptor (hPR), chicken progesterone receptor hinge region (cPR), and chicken progesterone receptor A/B domain (PR22), revealed that immunoreactivity was present in the epithelium of the olfactory organ of females and males of the trout Salmo trutta fario only against hPR. Western blotting showed two hPR immunoreactive bands of about 62 and 66 kDa. Finally, a portion of the cDNA of about 300 nucleotides extending over the DNA binding domain and the ligand binding domain was cloned and sequenced, revealing a high degree of sequence homology of the PR in Salmo trutta fario with the PR in other teleosts.
Collapse
Affiliation(s)
- Ettore Varricchio
- Department of Biological and Environmental Sciences, University of Sannio, Via Port'Arsa, 11, Benevento 82100, Italy
| | | | | | | |
Collapse
|
30
|
Hubert MA, Sherritt SL, Bachurski CJ, Handwerger S. Involvement of transcription factor NR2F2 in human trophoblast differentiation. PLoS One 2010; 5:e9417. [PMID: 20195529 PMCID: PMC2828470 DOI: 10.1371/journal.pone.0009417] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Accepted: 02/03/2010] [Indexed: 02/07/2023] Open
Abstract
Background During the in vitro differentiation of human villous cytotrophoblast (CTB) cells to a syncytiotrophoblast (STB) phenotype, mRNA levels for the nuclear hormone receptor NR2F2 (ARP-1, COUP-TFII) increase rapidly, reaching a peak at day 1 of differentiation that is 8.8-fold greater than that in undifferentiated CTB cells. To examine whether NR2F2 is involved in the regulation of villous CTB cell differentiation, studies were performed to determine whether NR2F2 regulates the expression of TFAP2A (AP-2α), a transcription factor that is critical for the terminal differentiation of these cells to a STB phenotype. Methodology/Primary Findings Overexpression of NR2F2 in primary cultures of human CTB cells and JEG-3 human choriocarcinoma cells induced dose-dependent increases in TFAP2A promoter activity. Conversely, siRNA mediated silencing of the NR2F2 gene in villous CTB undergoing spontaneous differentiation blocked the induction of the mRNAs for TFAP2A and several STB cell specific marker genes, including human placental lactogen (hPL), pregnancy specific glycoprotein 1 (PSG1) and corticotropin releasing hormone (CRH) by 51–59%. The induction of TFAP2A promoter activity by NR2F2 was potentiated by the nuclear hormone receptors retinoic acid receptor alpha (RARA) and retinoid X receptor alpha (RXRA). Conclusions/Significance Taken together, these results strongly suggest that NR2F2 is involved in villous CTB cell differentiation and that NR2F2 acts, at least in part, by directly activating TFAP2A gene expression and by potentiating the transactivation of TFAP2A by RARA and RXRA.
Collapse
Affiliation(s)
- Michael A. Hubert
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
- Division of Endocrinology, Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Susan L. Sherritt
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
- Division of Endocrinology, Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Cindy J. Bachurski
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
- Division of Pulmonary Biology, Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Stuart Handwerger
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
- Division of Endocrinology, Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
31
|
Abstract
Progesterone is a key hormone in the endometrium that opposes estrogen-driven growth. Insufficient progesterone will result in unopposed estrogen action that could lead to the development of endometrial hyperplasia and adenocarcinoma. Although these endometrial neoplasias can regress in response to progestin treatment, this does not occur in all instances. To understand this resistance to progesterone and to improve on existing hormonal therapies, it is imperative that the molecular mechanisms of progesterone action through its receptor be deciphered in endometrial cancer. This review highlights what is known thus far regarding the efficacy of progestin therapy in the clinic and the role of progesterone in endometrial cancer cell behavior and gene regulation.
Collapse
Affiliation(s)
- J Julie Kim
- Department Obstetrics and Gynecology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA.
| | | |
Collapse
|
32
|
Satterfield MC, Song G, Kochan KJ, Riggs PK, Simmons RM, Elsik CG, Adelson DL, Bazer FW, Zhou H, Spencer TE. Discovery of candidate genes and pathways in the endometrium regulating ovine blastocyst growth and conceptus elongation. Physiol Genomics 2009; 39:85-99. [DOI: 10.1152/physiolgenomics.00001.2009] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Establishment of pregnancy in ruminants requires blastocyst growth to form an elongated conceptus that produces interferon tau, the pregnancy recognition signal, and initiates implantation. Blastocyst growth and development requires secretions from the uterine endometrium. An early increase in circulating concentrations of progesterone (P4) stimulates blastocyst growth and elongation in ruminants. This study utilized sheep as a model to identify candidate genes and regulatory networks in the endometrium that govern preimplantation blastocyst growth and development. Ewes were treated daily with either P4 or corn oil vehicle from day 1.5 after mating to either day 9 or day 12 of pregnancy when endometrium was obtained by hysterectomy. Microarray analyses revealed many differentially expressed genes in the endometria affected by day of pregnancy and early P4 treatment. In situ hybridization analyses revealed that many differentially expressed genes were expressed in a cell-specific manner within the endometrium. The Database for Annotation, Visualization, and Integrated Discovery (DAVID) was used to identify functional groups of genes and biological processes in the endometrium that are associated with growth and development of preimplantation blastocysts. Notably, biological processes affected by day of pregnancy and/or early P4 treatment included lipid biosynthesis and metabolism, angiogenesis, transport, extracellular space, defense and inflammatory response, proteolysis, amino acid transport and metabolism, and hormone metabolism. This transcriptomic data provides novel insights into the biology of endometrial function and preimplantation blastocyst growth and development in sheep.
Collapse
Affiliation(s)
| | - Gwonhwa Song
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Kelli J. Kochan
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Penny K. Riggs
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Rebecca M. Simmons
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Christine G. Elsik
- Department of Biology, Georgetown University, Washington, District of Columbia
| | - David L. Adelson
- Department of Biology, Georgetown University, Washington, District of Columbia
| | - Fuller W. Bazer
- Centre for Bioinformatics and Computational Genetics, University of Adelaide, Australia; and
| | - Huaijun Zhou
- Department of Poultry Science, Texas A&M University, College Station, Texas
| | - Thomas E. Spencer
- Department of Animal Science, Texas A&M University, College Station, Texas
| |
Collapse
|
33
|
Baud O, Gressens P. Voie de signalisation Sonic Hedgehog et impact des glucocorticoïdes sur le cerveau en développement. Med Sci (Paris) 2009; 25:713-7. [DOI: 10.1051/medsci/2009258-9713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
34
|
Simon L, Spiewak KA, Ekman GC, Kim J, Lydon JP, Bagchi MK, Bagchi IC, DeMayo FJ, Cooke PS. Stromal progesterone receptors mediate induction of Indian Hedgehog (IHH) in uterine epithelium and its downstream targets in uterine stroma. Endocrinology 2009; 150:3871-6. [PMID: 19372202 PMCID: PMC2717869 DOI: 10.1210/en.2008-1691] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Uterine receptivity to embryo implantation depends on appropriate progesterone (P4) and estrogen stimulation. P4 rapidly stimulates production of the morphogen Indian hedgehog (IHH) in murine uterine epithelium as well as downstream molecules in the hedgehog pathway such as Patched homolog 1 (PTCH1) and nuclear receptor subfamily 2, group F, member 2 (NR2F2) in uterine stroma. Studies using IHH-null mice indicate that IHH is obligatory for the normal P4 response in the uterus. To determine whether IHH induction in uterine epithelium is mediated through P4 receptor (PR) in epithelium (E) and/or stroma (S), we produced tissue recombinants using uteri from neonatal PR knockout (ko) mice and wild-type (wt) mice containing PR in S and/or E or lacking PR altogether using a tissue recombinant methodology and assessed their response to P4. In tissue recombinants containing wt-S (wt-S + wt-E and wt-S + ko-E), P4 induced Ihh mRNA expression at 6 h that was 6-fold greater than in oil-treated controls (P < 0.05; n = 6) in both types of tissue recombinants despite the absence of epithelial PR in wt-S + ko-E grafts. Conversely, Ihh mRNA expression was unaffected by P4 in ko-S + ko-E and ko-S + wt-E grafts despite epithelial PR expression in the latter. Nr2f2 and Ptch1 mRNA expression was similar in that it was stimulated by P4 only in recombinants containing stromal PR. These results indicate that stromal PR is both necessary and sufficient for P4 stimulation of epithelial IHH as well as downstream events such as PTCH1 and NR2F2 increases in stroma.
Collapse
Affiliation(s)
- Liz Simon
- Department of Veterinary Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois 61802, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gulino A, De Smaele E, Ferretti E. Glucocorticoids and neonatal brain injury: the hedgehog connection. J Clin Invest 2009; 119:243-6. [PMID: 19244604 DOI: 10.1172/jci38387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Glucocorticoids (GCs) play a critical role in neural development; however, their prenatal or neonatal therapeutic use can have detrimental effects on the developing brain. In this issue of the JCI, Heine and Rowitch report that the molecular mechanisms underlying these detrimental effects involve the sonic hedgehog (Shh) signaling pathway, a crucial regulator of brain development and neural stem/progenitor cells (see the related study beginning on page 267). They show that GCs suppress Shh-induced proliferation of cerebellar progenitor cells in postnatal mice and that, conversely, Shh signaling is protective against GC-induced neonatal cerebellar injury by inducing the enzyme 11betaHSD2, which inactivates the GCs corticosterone and prednisolone, but not dexamethasone. The data provide a rationale for the therapeutic use of 11betaHSD2-sensitive GCs, but not dexamethasone, or for the exploitation of the neuroprotective effect of Shh agonists to prevent GC-induced pre- or neonatal brain injury.
Collapse
Affiliation(s)
- Alberto Gulino
- Department of Experimental Medicine, Sapienza University, Rome, Italy.
| | | | | |
Collapse
|
36
|
Pan Q, Chegini N. MicroRNA signature and regulatory functions in the endometrium during normal and disease states. Semin Reprod Med 2008; 26:479-93. [PMID: 18951330 DOI: 10.1055/s-0028-1096128] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
During the menstrual cycle, human endometrium undergoes extensive cyclic morphologic and biochemical modifications in preparation for embryo implantation. These processes are highly regulated by ovarian steroids and various locally expressed gene products and involve inflammatory reaction, apoptosis, cell proliferation, angiogenesis, differentiation (tissue formation), and tissue remodeling. MicroRNAs (miRNAs) have emerged as key regulators of gene expression, and their altered and/or aberrant expression has been associated with establishment and progression of various disorders, including tumorigenesis. This review highlights the endometrial expression of miRNAs and their potential regulatory functions under normal and pathologic conditions such as endometriosis, dysfunctional uterine bleeding, and endometrial cancer. Given the key regulatory function of miRNAs on gene expression stability, understanding the underlying mechanisms of how endometrial miRNAs are regulated and identifying their specific target genes and their functions might lead to the development of preventive and therapeutic strategies by regulating specific target genes associated with such reproductive disorders.
Collapse
Affiliation(s)
- Qun Pan
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Florida, Gainesville, Florida 32610, USA
| | | |
Collapse
|
37
|
Franco HL, Jeong JW, Tsai SY, Lydon JP, DeMayo FJ. In vivo analysis of progesterone receptor action in the uterus during embryo implantation. Semin Cell Dev Biol 2008; 19:178-86. [DOI: 10.1016/j.semcdb.2007.12.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Accepted: 12/29/2007] [Indexed: 11/17/2022]
|
38
|
Wakitani S, Hondo E, Phichitraslip T, Stewart CL, Kiso Y. Upregulation of Indian hedgehog gene in the uterine epithelium by leukemia inhibitory factor during mouse implantation. J Reprod Dev 2008; 54:113-6. [PMID: 18239353 DOI: 10.1262/jrd.19120] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leukemia inhibitory factor (LIF) and Indian hedgehog (Ihh) are essential for embryo implantation in mice and are regulated by the actions of 17beta-estradiol (E2) and progesterone, respectively. The present study examined the effect of LIF on Ihh and Ihh-related factors in the uterine luminal epithelium during the implantation period using a DNA microarray. Expression of Ihh mRNA reached its peak on the forth day of pregnancy, and progesterone receptor (Pgr) mRNA decreased on the fifth day of pregnancy in wildtype mice. On the other hand, these changes in expression were not seen in LIF-/- mice. Ihh and Pgr mRNA were upregulated by LIF injection in delayed implantation mice. This up-regulation of Pgr was transient and preceded an increase of Ihh mRNA. Ihh mRNA also increased after E2 injection in delayed implantation mice of the LIF-/- genotype. E2 did not affect transcription of Pgr mRNA in the uterine luminal epithelium of delayed implantation LIF-/- mice. Using an antibody against the C-terminal epitope of Ihh, unprocessed Ihh proteins, but not C-terminal peptides, by autoproteolytic cleavage of Ihh were detected by western blot analysis. Unprocessed Ihh did not show quantitative changes between the wildtype and LIF-/- mice during the implantation period. Transcription of hedgehog acyltransferase was not influenced by LIF and E2 injection. In conclusion, LIF, which has a crucial role in E2 action for initiation of implantation, caused transient induction of Pgr mRNA and subsequent upregulation of Ihh mRNA, which mediates progesterone-Pgr actions for successful implantation.
Collapse
Affiliation(s)
- Shoichi Wakitani
- Department of Veterinary Anatomy, Faculty of Agriculture, Yamaguchi University, Yamaguchi, Japan
| | | | | | | | | |
Collapse
|
39
|
Lee KY, Jeong JW, Tsai SY, Lydon JP, DeMayo FJ. Mouse models of implantation. Trends Endocrinol Metab 2007; 18:234-9. [PMID: 17588769 DOI: 10.1016/j.tem.2007.06.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 05/02/2007] [Accepted: 06/13/2007] [Indexed: 11/17/2022]
Abstract
The process of implantation, necessary for nearly all viviparous birth, consists of tightly regulated reactions including apposition of the blastocyst, attachment to the uterine epithelium and decidualization of the uterine stroma. In order for implantation to be successful, a reciprocal interaction between an implantation competent blastocyst and receptive uterus must be achieved. A more thorough understanding of the molecular mechanisms that regulate uterine receptivity and implantation is of clinical relevance to correct implantation failure and improve pregnancy rates. As molecular methodologies have evolved in recent times, the use of in vivo models to elucidate the molecular mechanisms involved in implantation has increased. The mouse has emerged as a powerful model to investigate implantation owing to the ability to control uterine physiology through exogenous stimuli, and more recently, the ability to manipulate gene expression. This review describes the evolution of the mouse as a model for understanding uterine implantation, including exciting new advances in this field, and describes a novel genetic pathway that can be elucidated from these models.
Collapse
Affiliation(s)
- Kevin Y Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
40
|
Thuróczy J, Reisvaag GJK, Perge E, Tibold A, Szilágyi J, Balogh L. Immunohistochemical Detection of Progesterone and Cellular Proliferation in Canine Mammary Tumours. J Comp Pathol 2007; 137:122-9. [PMID: 17645888 DOI: 10.1016/j.jcpa.2007.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Accepted: 05/27/2007] [Indexed: 11/22/2022]
Abstract
Immunohistochemical expression of progesterone and the cellular proliferation marker Ki-67 was assessed in formalin-fixed, paraffin wax-embedded samples from 43 canine mammary tumours. Benign tumours showed high surface membrane progesterone expression (mean 196.42+/-25.91 positively labelled cells per 500 tumour cells) and low cellular proliferation (52.14+/-16.73 positively labelled cells per 500 tumour cells), whilst malignant tumours had low progesterone expression (68.19+/-17.53 positively labelled cells per 500 tumour cells) and higher cellular proliferation (141.72+/-23.65 positively labelled cells per 500 tumour cells), the difference being statistically significant (P<0.005) in both cases. These findings suggest that the majority of progesterone receptors in canine mammary tumour tissue are not associated with bound progesterone. The progression towards malignancy in spontaneously arising canine mammary tumours therefore appears to be associated with a decrease in steroid hormone dependency. Progesterone expression was also noted in the cytoplasm of tumour cells where it may be associated with a cellular repair mechanism. This hypothesis is supported by the finding of significantly higher progesterone content in the cytoplasm of benign tumour cells.
Collapse
Affiliation(s)
- J Thuróczy
- Department of Obstetrics and Reproduction, Faculty of Veterinary Science, Szent István University, P.O. Box 2, H-1400 Budapest, Hungary.
| | | | | | | | | | | |
Collapse
|
41
|
Lewis MT, Visbal AP. The hedgehog signaling network, mammary stem cells, and breast cancer: connections and controversies. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2007:181-217. [PMID: 17939302 DOI: 10.1007/2789_2007_051] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Several signal transduction networks have been implicated in the regulation of mammary epithelial stem cell self-renewal and maintenance (Kalirai and Clarke 2006; Liu et al. 2005). These signaling networks include those of the Wnt, Notch, TGFO, EGF, FGF, IGF, and most recently, the Hedgehog (Hh) families of secreted ligands. However, we currently know very little about the cellular and molecular mechanisms by which these signaling pathways function to regulate normal epithelial stem/progenitor cells. What is clear is that the regulatory signaling networks thought to control normal stem/progenitor cell self-renewal and maintenance are, with the current sole exception of the hedgehog network, well-documented to have contributory roles in mammary cancer development and disease progression when misregulated. In this review, genetic regulation of mammary gland development by hedgehog network genes is outlined, highlighting a developing controversy as to whether activated hedgehog signaling regulates normal regenerative mammary epithelial stem cells or, indeed, whether activated hedgehog signaling functions at all in ductal development. In addition, the question of whether inappropriate hedgehog network activation influences breast cancer development is addressed, with emphasis on the prospects for using hedgehog signaling antagonists clinically for breast cancer treatment or prevention.
Collapse
Affiliation(s)
- M T Lewis
- Baylor Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Room N1210; MS:BCM600, One Baylor Plaza, 77030 Houston, TX, USA.
| | | |
Collapse
|