1
|
Alamoudi AJ, Alotaibi HT, Hareeri RH, Rizg WY, Abdel-Naim AB. Cordycepin alleviates endometrial hyperplasia in rats via alteration of PTEN/PI3K/Akt axis. J Funct Foods 2024; 119:106363. [DOI: 10.1016/j.jff.2024.106363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
|
2
|
Weber K, Bruer G, Krueger N, Schuster TB, Creutzenberg O, Schaudien D. Regenerative and progressing lesions in lungs and lung-associated lymph nodes from fourteen 90-day inhalation studies with chemically different particulate materials. Toxicol Lett 2024; 399 Suppl 1:49-72. [PMID: 38159619 DOI: 10.1016/j.toxlet.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 01/03/2024]
Abstract
Rat lungs and lung-associated lymph nodes from 14 inhalation studies with chemically different particulate materials were histopathologically re-evaluated, and the bronchoalveolar lavage fluid (BALF) data and lung burden analyses were compared. All investigated substances caused similar lesions. For most substances, 1 mg/m3 of respirable particulate matter was established as the borderline for adverse morphological changes after the 90-day exposure period, confirmed by the increase in polymorphonuclear neutrophils in BALF. Possible reversibility was demonstrated when recovery groups are included in the study especially allowing the differentiation between regeneration or progressing of inflammatory changes during the recovery period. It was concluded, that the major driver of toxicity is not an intrinsic chemical property of the particle but a particle effect. Concerning classification for specific target organ toxicant (STOT) repeated exposure (RE), this paper highlights that merely comparing the lowest concentration, at which adverse effects were observed, with the Classification Labelling and Packaging (CLP) regulation (EC) no. 1272/2008 guidance values is inappropriate and might lead to a STOT classification under CLP for a large part of the substances discussed in this paper, on the basis of typically mild to moderate findings in rat lung and lung-associated lymph nodes on day 1 after exposure. An in-depth evaluation of the pathologic findings is required and an expert judgement has to be included in the decision on classification and labeling, evaluating the type and severity of effects and comparing these with the classification criteria.
Collapse
Affiliation(s)
| | - Gustav Bruer
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Germany
| | - Nils Krueger
- Evonik Operations GmbH, Smart Materials, Hanau, Germany
| | | | - Otto Creutzenberg
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Germany.
| |
Collapse
|
3
|
Olowofolahan A, Fatunsin O, Olorunsogo O. Modulatory effect of ciprofloxacin, a broad spectrum antibacterial drug, on mPT pore using rat model with estradiol benzoate-induced endometrial hyperplasia. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3331-3341. [PMID: 37943297 DOI: 10.1007/s00210-023-02824-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023]
Abstract
Mitochondrial permeability transition (mPT) pore has become a motive for drug evolvement pertinent to dysregulated apoptosis situations. Some chemical compounds impede tumor/cancer via the inception of mPT pore opening. Ciprofloxacin has been demonstrated to hinder growth and effect apoptosis in some cancer cells. However, using a rat model, this study investigated its effect on mitochondrial-mediated cell death via mPT pore opening and estradiol benzoate (EB)-induced endometrial hyperplasia. Mitochondria were isolated using differential centrifugation. The opening of the pore, cytochrome c release (CCR), mitochondrial ATPase (mATPase) activity, mitochondrial lipid peroxidation (mLPO), caspases 3 and 9 levels, and hepatic DNA fragmentation were determined. Histological evaluation of hepatic and uterine sections and immunoexpression levels of Bax, caspase 3, and anti-apoptotic Bcl-2 levels were quantified. The results show that ciprofloxacin caused mPT pore opening, CCR, mATPase activity, effected mLPO, caspases 3 and 9 activations, and hepatic DNA fragmentation. The histology of the liver section showed moderate to marked disseminated congestion at 100 mg/kg, while higher doses showed severe hepatic damage. Severe EH was detected in the EB-treated rats which was attenuated by ciprofloxacin in the treatment group. The Bax and caspase expressions were upregulated by ciprofloxacin while anti-apoptotic Bcl-2 was downregulated. Ciprofloxacin induces mitochondrial-mediated cell death via mPT pore opening and mitigates EB-induced EH in rat models via Bax/caspase/Bcl-2 signaling pathway.
Collapse
Affiliation(s)
- Adeola Olowofolahan
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Omowumi Fatunsin
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunso Olorunsogo
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
4
|
Hsieh K, Bloom JR, Dickstein DR, Shah A, Yu C, Nehlsen AD, Resende Salgado L, Gupta V, Chadha M, Sindhu KK. Risk-Tailoring Radiotherapy for Endometrial Cancer: A Narrative Review. Cancers (Basel) 2024; 16:1346. [PMID: 38611024 PMCID: PMC11011021 DOI: 10.3390/cancers16071346] [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: 02/01/2024] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Endometrial cancer is the most common gynecologic cancer in the United States and it contributes to the second most gynecologic cancer-related deaths. With upfront surgery, the specific characteristics of both the patient and tumor allow for risk-tailored treatment algorithms including adjuvant radiotherapy and systemic therapy. In this narrative review, we discuss the current radiation treatment paradigm for endometrial cancer with an emphasis on various radiotherapy modalities, techniques, and dosing regimens. We then elaborate on how to tailor radiotherapy treatment courses in combination with other cancer-directed treatments, including chemotherapy and immunotherapy. In conclusion, this review summarizes ongoing research that aims to further individualize radiotherapy regimens for individuals in an attempt to improve patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kunal K. Sindhu
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
5
|
Wang H, Ma X, Jiang Z, Xia D, Sui F, Fu F, Dai Y. Estrogen promotes the proliferation and migration of endometrial cancer cells by upregulating the expression of lncRNA HOTAIR. Gynecol Endocrinol 2023; 39:2269248. [PMID: 37846544 DOI: 10.1080/09513590.2023.2269248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
OBJECTIVE Estrogen (E2) is the main contributor to the progression of endometrial cancer (EC). The long noncoding RNA HOX antisense intergenic RNA (HOTAIR) is emerging as a new regulator in several cancer types. This study aimed to investigate the role of HOTAIR in EC development and identify the underlying molecular mechanisms. METHODS HOTAIR expression levels in human EC tissues and the corresponding adjacent tissues and human EC Ishikawa cells were determined by quantitative PCR. Ishikawa cells were treated with E2 or estrogen receptor (ER) inhibitor ICI182780, transfected with siHOTAIR oligo, or infected with lentivirus expressing shHOTAIR/shNC, alone or in combinations. The protein expression of polycomb repressive complex 2 (PRC2) was evaluated by western blotting, and cell migration was measured by transwell assays. A xenograft tumorigenic model was established by inoculating control or stable shHOTAIR-infected Ishikawa cells into nude mice and implanting 17β-estradiol release pellets. RESULTS HOTAIR expression was significantly elevated in human EC tissues. E2 exposure markedly increased HOTAIR levels in Ishikawa cells. Notably, E2 increased the protein expression of PRC2 and promoted EC cell migration, which were dependent on HOTAIR expression, as HOTAIR knockdown abolished these effects of E2. Similarly, E2 promoted the in vivo proliferation of grafted Ishikawa cells via upregulated HOTAIR expression in nude mice. CONCLUSIONS Human EC tissues highly express HOTAIR, and E2-induced EC progression depends on HOTAIR expression. This work suggests that the E2-HOTAIR axis is a potential therapeutic target in EC therapy.
Collapse
Affiliation(s)
- Huixiao Wang
- Department of Maternal Intensive Care Unit, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xulan Ma
- Department of Gynecology, Aerospace Center Hospital, Beijing, China
| | - Ziwen Jiang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing,China
| | - Di Xia
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing,China
| | - Feng Sui
- Department of Maternal Intensive Care Unit, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fengxian Fu
- Department of Gynecology, Aerospace Center Hospital, Beijing, China
| | - Yinmei Dai
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing,China
| |
Collapse
|
6
|
Abdelzaher WY, Ibrahim MA, Hassan M, El-Tahawy NFG, Fawzy MA, Hafez HM. Protective effect of eicosapentaenoic acid against estradiol valerate-induced endometrial hyperplasia via modulation of NF-κB/HIF-1α/VEGF signaling pathway in rats. Chem Biol Interact 2023; 373:110399. [PMID: 36774993 DOI: 10.1016/j.cbi.2023.110399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023]
Abstract
BACKGROUND Early diagnosis and treatment of endometrial hyperplasia (EH) remains mandatory for endometrial cancer (EC) prevention. OBJECTIVE To study the possible protective effect of eicosapentaenoic acid (EPA) in EH - induced by estradiol valerate (EV) in rats. METHODS/MATERIALS Adult female Wistar rats were given EV with or without EPA for 10 days. The uterine changes were evaluated by both physical (weight index) and histopathological methods. The markers of oxidative stress (Uterine malondialdehyde (MDA) and serum total antioxidant capacity (TAC) as well as serum estradiol and progesterone levels, and apoptosis (uterine caspase-3) were determined. Immunohistochemical estimations of nuclear factor kappa B (NF-κB) and vascular endothelial growth factor (VEGF) in addition to hypoxia-inducible factor 1 alpha (HIF-1α) immunoblotting were measured in uterine tissue. KEY FINDINGS EV showed significant increase in uterine weight index that is accompanied with histopatholigical evidences of EH. Such changes were associated with significant alterations in oxidative stress markers, modulation of estradiol and progesterone serum levels, an increase in HIF-1α, NF-κB and VEGF immuno-expressions and a significant decrease in caspase-3. EPA, in either dose, showed significant amelioration in uterine weight index as well as in histopathological changes. Such effect was accompanied with significant improvement in the measured hormonal levels, oxidative stress, apoptosis, and inflammatory parameters. CONCLUSIONS EPA in the used doses provided biochemical and histopathological improvement in EV-induced EH via modulation of NF-κB/HIF-1α/VEGF signaling pathway.
Collapse
Affiliation(s)
| | - Mohamed A Ibrahim
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, 61511, Egypt.
| | - Marwa Hassan
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, 61511, Egypt.
| | | | - Michael Atef Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61511, Egypt.
| | - Heba M Hafez
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, 61511, Egypt.
| |
Collapse
|
7
|
Hernandez‐Jerez AF, Adriaanse P, Aldrich A, Berny P, Coja T, Duquesne S, Focks A, Millet M, Pelkonen O, Pieper S, Tiktak A, Topping CJ, Widenfalk A, Wilks M, Wolterink G, Angeli K, Recordati C, Van Durseen M, Aiassa E, Lanzoni A, Lostia A, Martino L, Guajardo IPM, Panzarea M, Terron A, Marinovich M. Development of adverse outcome pathways relevant for the identification of substances having endocrine disruption properties Uterine adenocarcinoma as adverse outcome. EFSA J 2023; 21:e07744. [PMID: 36818642 PMCID: PMC9926893 DOI: 10.2903/j.efsa.2023.7744] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Development of adverse outcome pathways (AOPs) for uterine adenocarcinoma can provide a practical tool to implement the EFSA-ECHA Guidance (2018) for the identification of endocrine disruptors in the context of Regulations (EU) No 528/2012 and (EC) No 1107/2009. AOPs can give indications about the strength of the relationship between an adverse outcome (intended as a human health outcome) and chemicals (pesticides but not only) affecting the pathways. In this scientific opinion, the PPR Panel explored the development of AOPs for uterine adenocarcinoma. An evidence-based approach methodology was applied, and literature reviews were produced using a structured framework assuring transparency, objectivity, and comprehensiveness. Several AOPs were developed; these converged to a common critical node, that is increased estradiol availability in the uterus followed by estrogen receptor activation in the endometrium; therefore, a putative AOP network was considered. An uncertainty analysis and a probabilistic quantification of the weight of evidence have been carried out via expert knowledge elicitation for each set of MIEs/KEs/KERs included in individual AOPs. The collected data on the AOP network were evaluated qualitatively, whereas a quantitative uncertainty analysis for weight of the AOP network certainty has not been performed. Recommendations are provided, including exploring further the uncertainties identified in the AOPs and putative AOP network; further methodological developments for quantifying the certainty of the KERs and of the overall AOPs and AOP network; and investigating of NAMs applications in the context of some of the MIEs/KEs currently part of the putative AOP network developed.
Collapse
|
8
|
Rush CM, Blanchard Z, Polaski JT, Osborne KS, Osby K, Vahrenkamp JM, Yang CH, Lum DH, Hagan CR, Leslie KK, Pufall MA, Thiel KW, Gertz J. Characterization of HCI-EC-23 a novel estrogen- and progesterone-responsive endometrial cancer cell line. Sci Rep 2022; 12:19731. [PMID: 36396974 PMCID: PMC9672046 DOI: 10.1038/s41598-022-24211-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Most endometrial cancers express the hormone receptor estrogen receptor alpha (ER) and are driven by excess estrogen signaling. However, evaluation of the estrogen response in endometrial cancer cells has been limited by the availability of hormonally responsive in vitro models, with one cell line, Ishikawa, being used in most studies. Here, we describe a novel, adherent endometrioid endometrial cancer (EEC) cell line model, HCI-EC-23. We show that HCI-EC-23 retains ER expression and that ER functionally responds to estrogen induction over a range of passages. We also demonstrate that this cell line retains paradoxical activation of ER by tamoxifen, which is also observed in Ishikawa and is consistent with clinical data. The mutational landscape shows that HCI-EC-23 is mutated at many of the commonly altered genes in EEC, has relatively few copy-number alterations, and is microsatellite instable high (MSI-high). In vitro proliferation of HCI-EC-23 is strongly reduced upon combination estrogen and progesterone treatment. HCI-EC-23 exhibits strong estrogen dependence for tumor growth in vivo and tumor size is reduced by combination estrogen and progesterone treatment. Molecular characterization of estrogen induction in HCI-EC-23 revealed hundreds of estrogen-responsive genes that significantly overlapped with those regulated in Ishikawa. Analysis of ER genome binding identified similar patterns in HCI-EC-23 and Ishikawa, although ER exhibited more bound sites in Ishikawa. This study demonstrates that HCI-EC-23 is an estrogen- and progesterone-responsive cell line model that can be used to study the hormonal aspects of endometrial cancer.
Collapse
Affiliation(s)
- Craig M Rush
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Zannel Blanchard
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jacob T Polaski
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kyle S Osborne
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Krystle Osby
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jeffery M Vahrenkamp
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chieh-Hsiang Yang
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - David H Lum
- Preclinical Research Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Christy R Hagan
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kimberly K Leslie
- Division of Molecular Medicine, Departments of Internal Medicine and Obstetrics and Gynecology, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Miles A Pufall
- Department of Biochemistry and Molecular Biology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kristina W Thiel
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, USA
| | - Jason Gertz
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
9
|
Gelissen JH, Huang GS. Intersections of endocrine pathways and the epithelial mesenchymal transition in endometrial cancer. Front Oncol 2022; 12:914405. [PMID: 36052252 PMCID: PMC9424890 DOI: 10.3389/fonc.2022.914405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
The epithelial mesenchymal transition (EMT) is the process by which cancer cells of epithelial origin, including endometrial cancer, acquire a mesenchymal phenotype with enhanced migratory and invasive capacity, to facilitate metastasis. The regulation of EMT is tissue-specific, and in endometrial cancer, endocrine signaling pathways serve as critical regulators of EMT. The intersections of endocrine signaling and EMT highlight potential avenues for therapeutic intervention to target cancer metastasis with the aim of reduced mortality.
Collapse
|
10
|
Benjamin K, Marquez CM, Morta M, Reyes EM, Aragones L, Velarde M. Bisphenol S Increases Cell Number and Stimulates Migration of Endometrial Epithelial Cells. J ASEAN Fed Endocr Soc 2022; 38:13-22. [PMID: 37234927 PMCID: PMC10207871 DOI: 10.15605/jafes.037.s7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/13/2022] [Indexed: 09/08/2023] Open
Abstract
OBJECTIVE To determine whether bisphenol S (BPS), a common substitute for bisphenol A (BPA), induces cell proliferation and migration in human endometrial epithelial cells (Ishikawa) and adult mouse uterine tissues. METHODOLOGY Human endometrial Ishikawa cells were exposed to low doses of BPS (1 nM and 100 nM) for 72 hours. Cell proliferation was assessed through the viability assays MTT and CellTiter-Glo®. Wound healing assays were also used to evaluate the migration potential of the cell line. The expression of genes related to proliferation and migration was also determined. Similarly, adult mice were exposed to BPS at a dose of 30 mg/kg body weight/day for 21 days, after which, the uterus was sent for histopathologic assessment. RESULTS BPS increased cell number and stimulated migration in Ishikawa cells, in association with the upregulation of estrogen receptor beta (ESR2) and vimentin (VIM). In addition, mice exposed to BPS showed a significantly higher mean number of endometrial glands within the endometrium. CONCLUSION Overall, in vitro and in vivo results obtained in this study showed that BPS could significantly promote endometrial epithelial cell proliferation and migration, a phenotype also observed with BPA exposure. Hence, the use of BPS in BPA-free products must be reassessed, as it may pose adverse reproductive health effects to humans.
Collapse
Affiliation(s)
- Kimberly Benjamin
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila
- Institute of Environmental Science and Meteorology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Cielo Mae Marquez
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Madeleine Morta
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Emmanuel Marc Reyes
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Lemnuel Aragones
- Institute of Environmental Science and Meteorology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Michael Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| |
Collapse
|
11
|
Eid BG. Chrysin attenuates estradiol-induced endometrial hyperplasia in rats via enhancing PPARα activity. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:54273-54281. [PMID: 35301625 DOI: 10.1007/s11356-022-19206-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
Endometrial hyperplasia (EH) is a complex condition that commonly affects women after menopause. Since the current available treatments for EH are mainly invasive, there is a need for developing new treatment modalities. Chrysin (Ch) is a dihydroxyflavone with numerous promising therapeutic potentials. In this study, Ch's protective effects on estradiol (E2)-induced EH were studied in rats. Animals were allocated randomly to five groups and were treated for 4 weeks as follows: Group 1, control: received the vehicle; group 2, Ch: received Ch 25 mg/kg; group 3, estradiol (E2): received E2 (3 mg/kg) 3 × weekly subcutaneously and the vehicle. Group 4, E2 + Ch 10 mg/kg and group 5, E2 + Ch 25 mg/kg: Ch was given once daily at 10 mg/kg or 25 mg/kg, respectively. In addition, E2 was administered 3 × weekly (3 mg/kg) in groups 4 and 5. Ch inhibited the E2-induced increase in uterine weights and histopathological changes. Ch lowered the cyclin D1 expression. Ch raised the caspase-3 content and Bax mRNA expression. Furthermore, it corrected the raised Bcl2 mRNA expression due to E2. Ch inhibited MDA accumulation and GSH depletion. It also prevents E2-induced SOD and GPx exhaustion. It also ameliorated the rise in NFκB, TNF-α, and IL-6 expression. These effects were correlated with an enhanced PPARα activity ratio relative to the E2 group. This suggests that Ch attenuates EH in this model by exerting anti-proliferative, anti-oxidant, and anti-inflammatory effects partially through increasing PPARα activity.
Collapse
Affiliation(s)
- Basma Ghazi Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
| |
Collapse
|
12
|
Ruan LY, Lai ZZ, Shi JW, Yang HL, Ye JF, Xie F, Qiu XM, Zhu XY, Li MQ. Excess Heme Promotes the Migration and Infiltration of Macrophages in Endometrial Hyperplasia Complicated with Abnormal Uterine Bleeding. Biomolecules 2022; 12:biom12060849. [PMID: 35740976 PMCID: PMC9221196 DOI: 10.3390/biom12060849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
In patients, endometrial hyperplasia (EH) is often accompanied by abnormal uterine bleeding (AUB), which is prone to release large amounts of heme. However, the role of excess heme in the migration and infiltration of immune cells in EH complicated by AUB remains unknown. In this study, 45 patients with AUB were divided into three groups: a proliferative phase group (n = 15), a secretory phase group (n = 15) and EH (n = 15). We observed that immune cell subpopulations were significantly different among the three groups, as demonstrated by flow cytometry analysis. Of note, there was a higher infiltration of total immune cells and macrophages in the endometrium of patients with EH. Heme up-regulated the expression of heme oxygenase-1 (HO-1) and nuclear factor erythroid-2-related factor 2 (Nrf2) in endometrial epithelial cells (EECs) in vitro, as well as chemokine (e.g., CCL2, CCL3, CCL5, CXCL8) levels. Additionally, stimulation with heme led to the increased recruitment of THP-1 cells in an indirect EEC-THP-1 co-culture unit. These data suggest that sustained and excessive heme in patients with AUB may recruit macrophages by increasing the levels of several chemokines, contributing to the accumulation and infiltration of macrophages in the endometrium of EH patients, and the key molecules of heme metabolism, HO-1 and Nrf2, are also involved in this regulatory process.
Collapse
Affiliation(s)
- Lu-Yu Ruan
- NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China;
- Laboratory for Reproductive Immunology, Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China; (Z.-Z.L.); (J.-W.S.); (H.-L.Y.)
| | - Zhen-Zhen Lai
- Laboratory for Reproductive Immunology, Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China; (Z.-Z.L.); (J.-W.S.); (H.-L.Y.)
| | - Jia-Wei Shi
- Laboratory for Reproductive Immunology, Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China; (Z.-Z.L.); (J.-W.S.); (H.-L.Y.)
| | - Hui-Li Yang
- Laboratory for Reproductive Immunology, Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China; (Z.-Z.L.); (J.-W.S.); (H.-L.Y.)
| | - Jiang-Feng Ye
- Institute for Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138632, Singapore;
| | - Feng Xie
- Medical Center of Diagnosis and Treatment for Cervical and Intrauterine Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- Correspondence: (F.X.); (X.-M.Q.); (X.-Y.Z.); (M.-Q.L.)
| | - Xue-Min Qiu
- Clinical Research Center, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Correspondence: (F.X.); (X.-M.Q.); (X.-Y.Z.); (M.-Q.L.)
| | - Xiao-Yong Zhu
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Correspondence: (F.X.); (X.-M.Q.); (X.-Y.Z.); (M.-Q.L.)
| | - Ming-Qing Li
- NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China;
- Laboratory for Reproductive Immunology, Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China; (Z.-Z.L.); (J.-W.S.); (H.-L.Y.)
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Correspondence: (F.X.); (X.-M.Q.); (X.-Y.Z.); (M.-Q.L.)
| |
Collapse
|
13
|
Binmahfouz LS, Eid BG, Bagher AM, Shaik RA, Binmahfouz NS, Abdel-Naim AB. Piceatannol SNEDDS Attenuates Estradiol-Induced Endometrial Hyperplasia in Rats by Modulation of NF-κB and Nrf2/HO-1 Axes. Nutrients 2022; 14:nu14091891. [PMID: 35565857 PMCID: PMC9102083 DOI: 10.3390/nu14091891] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 02/08/2023] Open
Abstract
Endometrial hyperplasia (EH) is the most common risk factor for endometrial malignancy in females. The pathogenesis of EH has been directly linked to uterine inflammation, which can result in abnormal cell division and decreased apoptosis. Piceatannol (PIC), a natural polyphenolic stilbene, is known to exert anti-inflammatory, antioxidant and anti-proliferative activities. The aim of the present study was to examine the potential preventive role of PIC in estradiol benzoate (EB)-induced EH in rats. A self-nanoemulsifying drug delivery system (SNEDDS) was prepared to improve the solubility of the PIC. Therefore, thirty female Wistar rats were divided into five groups: (1) control, (2) PIC SNEDDS (10 mg/kg), (3) EB (0.6 mg/kg), (4) EB + PIC SNEDDS (5 mg/kg) and (5) EB + PIC SNEDDS (10 mg/kg). The administration of PIC SNEDDS prevented EB-induced increases in uterine weights and histopathological changes. Additionally, it displayed pro-apoptotic and antioxidant activity in the endometrium. Immunohistochemical staining of uterine sections co-treated with PIC SNEDDS showed significantly decreased expression of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and nuclear transcription factor-kappa B (NF-κB). This anti-inflammatory effect was further confirmed by a significant increase in Nrf2 and heme oxygenase-1 (HO-1) expression. These results indicate that SNEDDS nanoformulation of PIC possesses protective effects against experimentally induced EH.
Collapse
Affiliation(s)
- Lenah S. Binmahfouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (B.G.E.); (A.M.B.); (R.A.S.); (A.B.A.-N.)
- Correspondence:
| | - Basma G. Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (B.G.E.); (A.M.B.); (R.A.S.); (A.B.A.-N.)
| | - Amina M. Bagher
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (B.G.E.); (A.M.B.); (R.A.S.); (A.B.A.-N.)
| | - Rasheed A. Shaik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (B.G.E.); (A.M.B.); (R.A.S.); (A.B.A.-N.)
| | - Najlaa S. Binmahfouz
- Department of Anatomical Histopathology, East Jeddah General Hospital, Jeddah 22253, Saudi Arabia;
| | - Ashraf B. Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (B.G.E.); (A.M.B.); (R.A.S.); (A.B.A.-N.)
| |
Collapse
|
14
|
Abruzzese GA, Silva AF, Velazquez ME, Ferrer MJ, Motta AB. Hyperandrogenism and Polycystic ovary syndrome: Effects in pregnancy and offspring development. WIREs Mech Dis 2022; 14:e1558. [PMID: 35475329 DOI: 10.1002/wsbm.1558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/18/2022] [Accepted: 04/01/2022] [Indexed: 11/10/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the major endocrine disorders affecting women of reproductive age. Its etiology remains unclear. It is suggested that environmental factors, and particularly the intrauterine environment, play key roles in PCOS development. Besides the role of androgens in PCOS pathogenesis, exposure to endocrine disruptors, as is Bisphenol A, could also contribute to its development. Although PCOS is considered one of the leading causes of ovarian infertility, many PCOS patients can get pregnant. Some of them by natural conception and others by assisted reproductive technique treatments. As hyperandrogenism (one of PCOS main features) affects ovarian and uterine functions, PCOS women, despite reaching pregnancy, could present high-risk pregnancies, including implantation failure, an increased risk of gestational diabetes, preeclampsia, and preterm birth. Moreover, hyperandrogenism may also be maintained in these women during pregnancy. Therefore, as an altered uterine milieu, including hormonal imbalance, could affect the developing organisms, monitoring these patients throughout pregnancy and their offspring development is highly relevant. The present review focuses on the impact of androgenism and PCOS on fertility issues and pregnancy-related outcomes and offspring development. The evidence suggests that the increased risk of pregnancy complications and adverse offspring outcomes of PCOS women would be due to the factors involved in the syndrome pathogenesis and the related co-morbidities. A better understanding of the involved mechanisms is still needed and could contribute to a better management of these women and their offspring. This article is categorized under: Reproductive System Diseases > Molecular and Cellular Physiology Reproductive System Diseases > Environmental Factors.
Collapse
Affiliation(s)
- Giselle A Abruzzese
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Aimé F Silva
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariela E Velazquez
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria-José Ferrer
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alicia B Motta
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
15
|
Jin Q, Jiang X, Du X, Hu W, Bai S, Wang X, Xu B, Zhao W. Integrated Transcriptome and Multiple Activated Pathways in Endometrial Cancer. Front Genet 2021; 12:680331. [PMID: 34925436 PMCID: PMC8678463 DOI: 10.3389/fgene.2021.680331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 10/07/2021] [Indexed: 12/05/2022] Open
Abstract
Because the incidence of endometrial cancer is notably increasing worldwide, it has become the leading gynecologic cancer in the United States. Standard treatment results in the loss of reproductive function in women of childbearing age. Furthermore, advanced cancer stages are associated with poor overall survival. The aim of this study was to explore the abnormal expression profile of genes during the development of endometrial cancer, which is essential to provide a better understanding of the mechanisms involved. Five pairs of endometrial cancer tissues and normal endometrial tissues were subjected to next-generation transcriptome sequencing technology. Quantitative real-time PCR (RT-qPCR) was performed to validate the expression profile of key differentially expressed genes (2.0-fold change, adj. p < 0.05) (DEGs) identified in the RNA-seq result. GO and KEGG pathways were used for bioinformatic analyses. The transcriptomic sequencing results showed 1153 DEGs, including 673 upregulated and 480 downregulated genes, in the EC specimens. Decreased expression of ID1, IGF1, GDF7, SMAD9, TGF-beta and WNT4, as well as GDF5, INHBA and ERBB4 overexpression, were confirmed in EC using RT-qPCR. Additionally, EC tissue exhibited marked enrichment in genes promoting cellular adhesion, proliferation, migration and plasma membrane. KEGG analysis revealed changes in various pathways, such as the TGF-beta, PI3K-Akt, Wnt, and estrogen pathways. Our data describe the molecular events involved in the pathogenesis of EC, which may be potential diagnostic markers and targets of therapeutic interventions.
Collapse
Affiliation(s)
- Qi Jin
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xiaohua Jiang
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xin Du
- Reproductive Medicine Center, 901th Hospital of PLA Joint Logistic Support Force, Hefei, China
| | - Weiping Hu
- Division of Life Sciences and Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Shun Bai
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xian Wang
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bo Xu
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- *Correspondence: Bo Xu, ; Weidong Zhao,
| | - Weidong Zhao
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Division of Life Sciences and Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- *Correspondence: Bo Xu, ; Weidong Zhao,
| |
Collapse
|
16
|
Hansda SR, Haldar C. Uterine anomalies in cell proliferation, energy homeostasis and oxidative stress in PCOS hamsters, M. auratus: Therapeutic potentials of melatonin. Life Sci 2021; 281:119755. [PMID: 34175318 DOI: 10.1016/j.lfs.2021.119755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/07/2021] [Accepted: 06/17/2021] [Indexed: 12/22/2022]
Abstract
AIMS Polycystic ovarian syndrome (PCOS) is a reproductive, endocrine and metabolic disorder. Less is known about the mechanism of its effect on uterine function and therapeutic potential of melatonin. Our aim was to evaluate uterine dysfunction(s) in letrozole induced PCOS and its possible rectification by melatonin. MAIN METHODS Adult female golden hamsters were divided into groups of Control (C), Melatonin (M; 1 mg/kg b.w.), Letrozole (L; 3 mg/kg b.w.) and combination of Letrozole+Melatonin (L + M; 3 mg/kg b.w. + 1 mg/kg b.w.) which were treated for 40 days. Analysis of serum testosterone/estradiol/progesterone/leptin/insulin, uterine histomorphometry, immunohistochemistry for proliferation cell nuclear antigen (PCNA), homeostatic assessment model of insulin resistance (HOMA-IR), western blotting for PCNA, androgen receptor (AR), insulin receptor (InsR), glucose tansporter-4 (GLUT-4), nuclear factor-kappa B (NFκB), cyclooxygenase-2 (COX-2) and biochemical analysis of superoxide dismutase (SOD)/catalase/lipid peroxidation (LPO) were done. KEY FINDINGS Serum testosterone, leptin and insulin increased while uterine InsR/GLUT-4 expression decreased in L group indicating metabolic abnormalities. Endometrial hyperplasia, increased expression of PCNA and AR indicated abnormal proliferation in L compared to C. Increased uterine oxidative load (SOD/catalase/LPO) and inflammatory markers NFκB/COX-2 expression in L was responsible for high tissue oxidative stress and inflammation. M administration normalized all the above parameters suggesting its ameliorative effect in L + M group. SIGNIFICANCE We report PCOS induced uterine dysfunction in Mesocricetus auratus for the first time. M administration restores uterine functions modulating cellular dynamicity, metabolic status, decreased oxidative and inflammatory load in PCOS hamsters. Therefore, we suggest the therapeutic potential of M against PCOS led uterine abnormalities to restore female fertility.
Collapse
Affiliation(s)
- Shruti R Hansda
- Pineal Research Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India.
| | - Chandana Haldar
- Pineal Research Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India.
| |
Collapse
|
17
|
Prolonged atrazine exposure beginning in utero and adult uterine morphology in mice. J Dev Orig Health Dis 2021; 13:39-48. [PMID: 33781367 DOI: 10.1017/s2040174421000106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Through drinking water, humans are commonly exposed to atrazine, a herbicide that acts as an endocrine and metabolic disruptor. It interferes with steroidogenesis, including promoting oestrogen production and altering cell metabolism. However, its precise impact on uterine development remains unknown. This study aimed to determine the effect of prolonged atrazine exposure on the uterus. Pregnant mice (n = 5/group) received 5 mg/kg body weight/day atrazine or DMSO in drinking water from gestational day 9.5 until weaning. Offspring continued to be exposed until 3 or 6 months of age (n = 5-9/group), when uteri were collected for morphological and molecular analyses and steroid quantification. Endometrial hyperplasia and leiomyoma were evident in the uteri of atrazine-exposed mice. Uterine oestrogen concentration, oestrogen receptor expression, and localisation were similar between groups, at both ages (P > 0.1). The expression and localisation of key epithelial-to-mesenchymal transition (EMT) genes and proteins, critical for tumourigenesis, remained unchanged between treatments, at both ages (P > 0.1). Hence, oestrogen-mediated changes to established EMT markers do not appear to underlie abnormal uterine morphology evident in atrazine exposure mice. This is the first report of abnormal uterine morphology following prolonged atrazine exposure starting in utero, it is likely that the abnormalities identified would negatively affect female fertility, although mechanisms remain unknown and require further study.
Collapse
|
18
|
Basappa B, Chumadathil Pookunoth B, Shinduvalli Kempasiddegowda M, Knchugarakoppal Subbegowda R, Lobie PE, Pandey V. Novel Biphenyl Amines Inhibit Oestrogen Receptor (ER)-α in ER-Positive Mammary Carcinoma Cells. Molecules 2021; 26:783. [PMID: 33546391 PMCID: PMC7913524 DOI: 10.3390/molecules26040783] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/20/2021] [Accepted: 01/23/2021] [Indexed: 11/17/2022] Open
Abstract
Herein, the activity of adamantanyl-tethered-biphenyl amines (ATBAs) as oestrogen receptor alpha (ERα) modulating ligands is reported. Using an ERα competitor assay it was demonstrated that ATBA compound 3-(adamantan-1-yl)-4-methoxy-N-(4-(trifluoromethyl) phenyl) aniline (AMTA) exhibited an inhibitory concentration 50% (IC50) value of 62.84 nM and demonstrated better binding affinity compared to tamoxifen (IC50 = 79.48 nM). Treatment of ERα positive (ER+) mammary carcinoma (MC) cells (Michigan Cancer Foundation-7 (MCF7)) with AMTA significantly decreased cell viability at an IC50 value of 6.4 μM. AMTA treatment of MC cell-generated three-dimensional (3D) spheroids resulted in significantly decreased cell viability. AMTA demonstrated a superior inhibitory effect compared to tamoxifen-treated MC cell spheroids. Subsequently, by use of an oestrogen response element (ERE) luciferase reporter construct, it was demonstrated that AMTA treatment significantly deceased ERE transcriptional activity in MC cells. Concordantly, AMTA treatment of MC cells also significantly decreased protein levels of oestrogen-regulated CCND1 in a dose-dependent manner. In silico molecular docking analysis suggested that AMTA compounds interact with the ligand-binding domain of ERα compared to the co-crystal ligand, 5-(4-hydroxyphenoxy)-6-(3-hydroxyphenyl)-7- methylnaphthalen-2-ol. Therefore, an analogue of AMTA may provide a structural basis to develop a newer class of ERα partial agonists.
Collapse
Affiliation(s)
- Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India;
- Department of Chemistry, Bangalore University, Bangalore 560001, India;
| | | | | | | | - Peter E. Lobie
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Beijing 518055, China;
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Beijing 518055, China
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Beijing 518055, China;
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Beijing 518055, China
| |
Collapse
|
19
|
Targeting Aquaporins in Novel Therapies for Male and Female Breast and Reproductive Cancers. Cells 2021; 10:cells10020215. [PMID: 33499000 PMCID: PMC7911300 DOI: 10.3390/cells10020215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/24/2022] Open
Abstract
Aquaporins are membrane channels in the broad family of major intrinsic proteins (MIPs), with 13 classes showing tissue-specific distributions in humans. As key physiological modulators of water and solute homeostasis, mutations, and dysfunctions involving aquaporins have been associated with pathologies in all major organs. Increases in aquaporin expression are associated with greater severity of many cancers, particularly in augmenting motility and invasiveness for example in colon cancers and glioblastoma. However, potential roles of altered aquaporin (AQP) function in reproductive cancers have been understudied to date. Published work reviewed here shows distinct classes aquaporin have differential roles in mediating cancer metastasis, angiogenesis, and resistance to apoptosis. Known mechanisms of action of AQPs in other tissues are proving relevant to understanding reproductive cancers. Emerging patterns show AQPs 1, 3, and 5 in particular are highly expressed in breast, endometrial, and ovarian cancers, consistent with their gene regulation by estrogen response elements, and AQPs 3 and 9 in particular are linked with prostate cancer. Continuing work is defining avenues for pharmacological targeting of aquaporins as potential therapies to reduce female and male reproductive cancer cell growth and invasiveness.
Collapse
|
20
|
Olowofolahan AO, Oyebode OT, Olorunsogo OO. Methyl palmitate reversed estradiol benzoate-induced endometrial hyperplasia in female rats. Toxicol Mech Methods 2021; 31:43-52. [PMID: 32967526 DOI: 10.1080/15376516.2020.1827329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/16/2020] [Accepted: 09/19/2020] [Indexed: 02/08/2023]
Abstract
Early detection and treatment of endometrial hyperplasia (EH) is mandatory for endometrial cancer prevention. Several bioactive agents of plant origin have been shown to elicit their chemotherapeutic effect against tumors and cancer via induction of mitochondrial permeability transition(mPT) pore opening. This research was therefore aimed at evaluating the potential chemopreventive effect of methyl palmitate (MP), on estradiol benzoate(EB)-induced EH, looking at the mitochondrial-mediated pathway and other possible mechanisms of action. Mitochondria were isolated using differential centrifugation. The mPT pore, mitochondrial ATPase (mATPase) activity, lipid peroxidation and cytochrome c release were determined by standard methods using spectrophotometer. Uterine interleukin 1b, MDA levels and SOD, GSH activities, were determined using commercially available kits. The uterine histological and immunohistochemical assessment of estrogen receptor (ERα), IL-1b and caspas-3 were carried out. The fibroblast cell count density was determined using histomorphometry. At all the concentrations of MP used, there was no significant induction of mPT pore opening, neither any enhancement of mATPase activity nor release of cytochrome c when compared to the control. Similar pattern of results were recorded for the in vivo study. However, there was marked increase in the uterine MDA and interleukin 1b levels, with concurrent decrease in SOD and GSH activities, in the EB-treated group, which was significantly reversed by MP co-administration. Endometrial Hyperplasia observed in the EB-treated group was ameliorated by MP co-administration. The immunoexpression of ERα and IL-1b in the EB-treated group was reversed by MP co-administration. This study suggests anti-inflammatory, antioxidant and anti-proliferative potential of MP against EB-induced EH.
Collapse
Affiliation(s)
- Adeola O Olowofolahan
- Laboratory for Membrane Biochemistry Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olubukola T Oyebode
- Laboratory for Membrane Biochemistry Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunso O Olorunsogo
- Laboratory for Membrane Biochemistry Research and Biotechnology, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
21
|
Singh S, Pavuluri S, Jyothi Lakshmi B, Biswa BB, Venkatachalam B, Tripura C, Kumar S. Molecular characterization of Wdr13 knockout female mice uteri: a model for human endometrial hyperplasia. Sci Rep 2020; 10:14621. [PMID: 32883989 PMCID: PMC7471898 DOI: 10.1038/s41598-020-70773-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 07/20/2020] [Indexed: 01/29/2023] Open
Abstract
Endometrial hyperplasia (EH) is a condition where uterine endometrial glands show excessive proliferation of epithelial cells that may subsequently progress into endometrial cancer (EC). Modern lifestyle disorders such as obesity, hormonal changes and hyperinsulinemia are known risk factors for EH. A mouse strain that mimics most of these risk factors would be an ideal model to study the stage-wise progression of EH disease and develop suitable treatment strategies. Wdr13, an X-linked gene, is evolutionarily conserved and expressed in several tissues including uteri. In the present study, Wdr13 knockout female mice developed benign proliferative epithelium that progressed into EH at around one year of age accompanied by an increase in body weight and elevated estradiol levels. Molecular characterization studies revealed increase in ERα, PI3K and a decrease in PAX2 and ERβ proteins in Wdr13 mutant mice uteri. Further, a decrease in the mRNA levels of cell cycle inhibitors, namely; p21 and cyclin G2 was seen. Leukocyte infiltration was observed in the uterine tissue of knockout mice at around 12 months of age. These physiological, molecular and pathological patterns were similar to those routinely seen in human EH disease and demonstrated the importance of WDR13 in mice uterine tissue. Thus, the genetic loss of Wdr13 in these mice led to mimicking of the human EH associated metabolic disorders making Wdr13 knockout female mice a potential animal model to study human endometrial hyperplasia.
Collapse
Affiliation(s)
- Shalu Singh
- Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Sivapriya Pavuluri
- Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, 500007, India
| | - B Jyothi Lakshmi
- Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Bhim B Biswa
- Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Bharathi Venkatachalam
- Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Chaturvedula Tripura
- Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Satish Kumar
- Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, 500007, India.
- Department of Biotechnology, School of Interdisciplinary and Applied Sciences, Central University of Haryana, Mahendergarh, Haryana, 123031, India.
| |
Collapse
|
22
|
Lai ZZ, Ruan LY, Wang Y, Yang HL, Shi JW, Wu JN, Qiu XM, Ha SY, Shen HH, Yang SL, Zheng ZM, Shao J, Ye JF, Li MQ. Changes in subsets of immunocytes in endometrial hyperplasia. Am J Reprod Immunol 2020; 84:e13295. [PMID: 32583503 DOI: 10.1111/aji.13295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
PROBLEM Endometrial hyperplasia (EH) is characterized by an endometrial gland-to-stroma ratio >1 and is one of the most common gynecological diseases in the world. The role of immunocyte subsets in the development of EH remains unknown. METHODS Patients who underwent dilatation and curettage due to abnormal uterine bleeding were recruited in the present study. Alterations in the numbers of different types of immune cell subsets in the endometrium of patients were analyzed by flow cytometry. RESULTS The present study included 48 patients who were divided into three groups, based on the pathological results: (a) proliferative period (PP, n = 12); (b) simple EH (SEH, n = 30); and (c) complex EH (CEH, n = 6). The results showed that immune cell subpopulations were significantly different between these three groups. Compared with the PP group, the proportion of CD45+ cells and neutrophils and the subtypes of T cells and macrophages were significantly increased in the SEH patients. Compared with the PP and SEH groups, subsets of immunocytes in the CEH group were significantly decreased, including the population of CD45+ cells and the subtypes of T cells and natural killer cells; in contrast, the proportion of macrophages was significantly increased. There were no significant differences between the other cell subsets in each group. CONCLUSION The changes in immune cell subsets may be closely associated with the progression of EH. Although the specific role of different immune cell subsets in the development of the diseases requires further study, the changes in the proportions of immune cell subsets should not be ignored.
Collapse
Affiliation(s)
- Zhen-Zhen Lai
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Lu-Yu Ruan
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Yan Wang
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Hui-Li Yang
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jia-Wei Shi
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jiang-Nan Wu
- Clinical Epidemiology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Xue-Min Qiu
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Si-Yao Ha
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Hui-Hui Shen
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Shao-Liang Yang
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Zi-Meng Zheng
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jun Shao
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jiang-Feng Ye
- Division of Obstetrics and Gynecology, KK Women's and Children's Hospital, Singapore City, Singapore
| | - Ming-Qing Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Liu Y, Zhao R, Chi S, Zhang W, Xiao C, Zhou X, Zhao Y, Wang H. UBE2C Is Upregulated by Estrogen and Promotes Epithelial-Mesenchymal Transition via p53 in Endometrial Cancer. Mol Cancer Res 2019; 18:204-215. [PMID: 31662448 DOI: 10.1158/1541-7786.mcr-19-0561] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 09/17/2019] [Accepted: 10/25/2019] [Indexed: 12/24/2022]
Abstract
Ubiquitin-conjugating enzyme E2C (UBE2C) plays important roles in tumor progression; nevertheless, its function in endometrial cancer remains unclear. This study elucidated the impact of UBE2C on endometrial cancer and its underlying mechanism. Human endometrial cancer and normal endometrial tissues were acquired from patients at Wuhan Union Hospital and UBE2C expression was detected by Western blotting and qRT-PCR. Endometrial cancer cells were transfected with a UBE2C overexpression plasmid or UBE2C-specific short hairpin RNA (shRNA) to up- or downregulate UBE2C expression, respectively. CCK8 and transwell assays were applied to assess the effects of UBE2C on cell proliferation, migration, and invasion. We found a significant elevation of UBE2C expression in patients with endometrial cancer, and that UBE2C upregulation was associated with advanced histologic grade, FIGO stage, recurrence, and shorter overall survival. UBE2C knockdown inhibited endometrial cancer cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), whereas UBE2C overexpression exerted the opposite effects. UBE2C downregulation increased p53 and its downstream p21 expression, with p53 overexpression reversing the EMT-promoting effects of UBE2C. UBE2C enhanced p53 ubiquitination to facilitate its degradation in endometrial cancer cells. Estradiol (E2) induced UBE2C expression via estrogen receptor α, which binds directly to the UBE2C promoter element. Silencing of UBE2C inhibited E2-promoted migration, invasion, and EMT in vitro and in vivo. IMPLICATIONS: UBE2C-mediated tumor EMT promotion by estrogen is a novel mechanism for the progression of estrogen-induced endometrial cancer, which could offer new biomarkers for diagnosis and therapy of endometrial cancer in the future.
Collapse
Affiliation(s)
- Yan Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rong Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuqi Chi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chengyu Xiao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xing Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yingchao Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
24
|
Ali FF, Abdelzaher WY, Ibrahim RA, Elroby Ali DM. Amelioration of estrogen-induced endometrial hyperplasia in female rats by hemin via heme-oxygenase-1 expression, suppression of iNOS, p38 MAPK, and Ki67. Can J Physiol Pharmacol 2019; 97:1159-1168. [PMID: 31505119 DOI: 10.1139/cjpp-2019-0287] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although heme oxygenase-1 (HO-1) is part of an endogenous defense system implicated in the homeostatic response, its role in cell proliferation and tumor progression is still controversial. Endometrial hyperplasia (EH) is associated with high risk of endometrial cancer (EC). Therefore, we aimed to evaluate the effect of hemin, a HO-1 inducer, against EH. Thirty-two female rats (60-70 days old) were divided into 4 groups treated for 1 week: vehicle control group, hemin group (25 mg/kg; i.p. 3 times/week), estradiol valerate (EV) group (2 mg/kg per day, p.o.), and hemin plus EV group. Sera were obtained for reduced glutathione level. Uterine malondialdehyde, superoxide dismutase, total nitrite/nitrate, and interleukin-1β levels were estimated. HO-1 and p38 mitogen-activated protein kinase expressions were obtained in uterine tissue. Uterine histological and immunohistochemical assessment of iNOS and Ki67 were also done. Results demonstrated that upregulation of HO-1 expression in hemin plus EV rats led to amelioration of EH which was confirmed with histological examination. This was associated with significant decrease in oxidative stress parameters, p38 mitogen-activated protein kinase expression, and interleukin-1β level. Also, uterine iNOS and Ki67 expressions were markedly suppressed. In conclusion, upregulation of HO-1 expression via hemin has ameliorative effect against EH through its antioxidant, anti-inflammatory, and antiproliferative actions.
Collapse
Affiliation(s)
- Fatma F Ali
- Department of Medical Physiology, Faculty of Medicine, Minia University, Egypt
| | | | - Randa Ahmed Ibrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Egypt
| | | |
Collapse
|
25
|
Raffone A, Travaglino A, Saccone G, Mollo A, De Placido G, Insabato L, Zullo F. Should progesterone and estrogen receptors be assessed for predicting the response to conservative treatment of endometrial hyperplasia and cancer? A systematic review and meta‐analysis. Acta Obstet Gynecol Scand 2019; 98:976-987. [DOI: 10.1111/aogs.13586] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 02/13/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Antonio Raffone
- Obstetrics and Gynecology Unit Department of Neuroscience, Reproductive Sciences and Dentistry School of MedicineUniversity of Naples Federico II Naples Italy
| | - Antonio Travaglino
- Anatomic Pathology Unit Department of Advanced Biomedical Sciences School of Medicine University of Naples Federico II Naples Italy
| | - Gabriele Saccone
- Obstetrics and Gynecology Unit Department of Neuroscience, Reproductive Sciences and Dentistry School of MedicineUniversity of Naples Federico II Naples Italy
| | - Antonio Mollo
- Obstetrics and Gynecology Unit Department of Neuroscience, Reproductive Sciences and Dentistry School of MedicineUniversity of Naples Federico II Naples Italy
| | - Giuseppe De Placido
- Obstetrics and Gynecology Unit Department of Neuroscience, Reproductive Sciences and Dentistry School of MedicineUniversity of Naples Federico II Naples Italy
| | - Luigi Insabato
- Anatomic Pathology Unit Department of Advanced Biomedical Sciences School of Medicine University of Naples Federico II Naples Italy
| | - Fulvio Zullo
- Obstetrics and Gynecology Unit Department of Neuroscience, Reproductive Sciences and Dentistry School of MedicineUniversity of Naples Federico II Naples Italy
| |
Collapse
|
26
|
Goad J, Ko YA, Kumar M, Jamaluddin MFB, Tanwar PS. Oestrogen fuels the growth of endometrial hyperplastic lesions initiated by overactive Wnt/β-catenin signalling. Carcinogenesis 2019; 39:1105-1116. [PMID: 29912292 DOI: 10.1093/carcin/bgy079] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
Unopposed oestrogen is responsible for approximately 80% of all the endometrial cancers. The relationship between unopposed oestrogen and endometrial cancer was indicated by the increase in the number of endometrial cancer cases due to the widespread use of oestrogen replacement therapy. Approximately 30% of the endometrial cancer patients have mutations in the Wnt signalling pathway. How the unbalanced ratios of ovarian hormones and the mutations in Wnt signalling pathway interact to cause endometrial cancer is currently unclear. To study this, we have developed a uterine epithelial cell-specific inducible cre mouse model and used 3D in vitro culture of human endometrial cancer cell lines. We showed that activating mutations in the Wnt signalling pathway for a prolonged period leads to endometrial hyperplasia but not endometrial cancer. Interestingly, unopposed oestrogen and activating mutations in Wnt signalling together drive the progression of endometrial hyperplasia to endometrial cancer. We have provided evidence that progesterone can be used as a targeted therapy against endometrial cancer cases presented with the activating mutations in Wnt signalling pathway.
Collapse
Affiliation(s)
- Jyoti Goad
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales Australia
| | - Yi-An Ko
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales Australia
| | - Manish Kumar
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales Australia
| | - M Fairuz B Jamaluddin
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales Australia
| | - Pradeep S Tanwar
- Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales Australia
| |
Collapse
|
27
|
Rodriguez AC, Blanchard Z, Maurer KA, Gertz J. Estrogen Signaling in Endometrial Cancer: a Key Oncogenic Pathway with Several Open Questions. Discov Oncol 2019; 10:51-63. [PMID: 30712080 PMCID: PMC6542701 DOI: 10.1007/s12672-019-0358-9] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/16/2019] [Indexed: 01/10/2023] Open
Abstract
Endometrial cancer is the most common gynecological cancer in the developed world, and it is one of the few cancer types that is becoming more prevalent and leading to more deaths in the USA each year. The majority of endometrial tumors are considered to be hormonally driven, where estrogen signaling through estrogen receptor α (ER) acts as an oncogenic signal. The major risk factors and some treatment options for endometrial cancer patients emphasize a key role for estrogen signaling in the disease. Despite the strong connections between estrogen signaling and endometrial cancer, important molecular aspects of ER function remain poorly understood; however, progress is being made in our understanding of estrogen signaling in endometrial cancer. Here, we discuss the evidence for the importance of estrogen signaling in endometrial cancer, details of the endometrial cancer-specific actions of ER, and open questions surrounding estrogen signaling in endometrial cancer.
Collapse
Affiliation(s)
- Adriana C Rodriguez
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Zannel Blanchard
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kathryn A Maurer
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jason Gertz
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA. .,Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA.
| |
Collapse
|
28
|
Cheal SM, Xu H, Guo HF, Patel M, Punzalan B, Fung EK, Lee SG, Bell M, Singh M, Jungbluth AA, Zanzonico PB, Piersigilli A, Larson SM, Cheung NKV. Theranostic pretargeted radioimmunotherapy of internalizing solid tumor antigens in human tumor xenografts in mice: Curative treatment of HER2-positive breast carcinoma. Am J Cancer Res 2018; 8:5106-5125. [PMID: 30429889 PMCID: PMC6217068 DOI: 10.7150/thno.26585] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022] Open
Abstract
In recent reports, we have shown that optimized pretargeted radioimmunotherapy (PRIT) based on molecularly engineered antibody conjugates and 177Lu-DOTA chelate (DOTA-PRIT) can be used to cure mice bearing human solid tumor xenografts using antitumor antibodies to minimally internalizing membrane antigens, GPA33 (colon) and GD2 (neuroblastoma). However, many solid tumor membrane antigens are internalized after antibody binding and it is generally believed that internalizing tumor membrane antigens are not suitable targets for PRIT. In this study, we tested the hypothesis that DOTA-PRIT can be performed successfully to target HER2, an internalizing membrane antigen widely expressed in breast, ovarian, and gastroesophageal junction cancers. Methods: DOTA-PRIT was carried out in athymic nude mice bearing BT-474 xenografts, a HER2-expressing human breast cancer, using a three-step dosing regimen consisting of sequential intravenous administrations of: 1) a bispecific IgG-scFv (210 kD) format (BsAb) carrying the IgG sequence of the anti-HER2 antibody trastuzumab and the scFv “C825” with high-affinity, hapten-binding antibody for Bn-DOTA (metal) (BsAb: anti-HER2-C825), 2) a 500 kD dextran-based clearing agent, followed by 3) 177Lu-DOTA-Bn. At the time of treatment, athymic nude mice bearing established subcutaneous BT-474 tumors (medium- and smaller-sized tumors with tumor volumes of 209 ± 101 mm3 and ranging from palpable to 30 mm3, respectively), were studied along with controls. We studied single- and multi-dose regimens. For groups receiving fractionated treatment, we verified quantitative tumor targeting during each treatment cycle using non-invasive imaging with single-photon emission computed tomography/computed tomography (SPECT/CT). Results: We achieved high therapeutic indices (TI, the ratio of radiation-absorbed dose in tumor to radiation-absorbed dose to critical organs, such as bone marrow) for targeting in blood (TI = 28) and kidney (TI = 7), while delivering average radiation-absorbed doses of 39.9 cGy/MBq to tumor. Based on dosimetry estimates, we implemented a curative fractionated therapeutic regimen for medium-sized tumors that would deliver approximately 70 Gy to tumors, which required treatment with a total of 167 MBq 177Lu-DOTA-Bn/mouse (estimated absorbed tumor dose: 66 Gy). This regimen was well tolerated and achieved 100% complete responses (CRs; defined herein as tumor volume equal to or smaller than 4.2 mm3), including 62.5% histologic cure (5/8) and 37.5% microscopic residual disease (3/8) at 85 days (d). Treatment controls showed tumor progression to 207 ± 201% of pre-treatment volume at 85 d and no CRs. Finally, we show that treatment with this curative 177Lu regimen leads to a very low incidence of histopathologic abnormalities in critical organs such as bone marrow and kidney among survivors compared with non-treated controls. Conclusion: Contrary to popular belief, we demonstrate that DOTA-PRIT can be successfully adapted to an internalizing antigen-antibody system such as HER2, with sufficient TIs and absorbed tumor doses to achieve a high probability of cures of established human breast cancer xenografts while sparing critical organs of significant radiotoxicity.
Collapse
|
29
|
Mechanisms underlying the protective effect of montelukast in prevention of endometrial hyperplasia in female rats. Int Immunopharmacol 2018; 62:326-333. [PMID: 30056375 DOI: 10.1016/j.intimp.2018.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 05/09/2018] [Accepted: 07/10/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To study the possible protective role of montelukast in endometrial hyperplesia (EH) rat model, induced by estradiol valerate (EV). METHODS/MATERIALS Thirty six female albino Wistar rats were classified into 7 groups: normal control, EV (2 mg/kg/day, p.o.), montelukast (10 mg/kg/day, p.o.), montelukast (1 mg/kg/day, p.o.) + EV (2 mg/kg/day, p.o.), montelukast (10 mg/kg/day, p.o.) + EV (2 mg/kg/day, p.o.), montelukast (20 mg/kg/day, p.o.) + EV (2 mg/kg/day, p.o.) groups. Uterine malondialdehyde (MDA), superoxide dismutase (SOD), total nitrites (NO) and serum total antioxidant capacity (TAC) were determined. Uterine, serum total cholesterol, high density lipoprotein (HDL) and tumor necrosis factor (TNF)-α were measured. Histopathological examination of the uterine tissue was also done. In addition, immunohistochemistry was done using Phosphatase and tensin homolog (PTEN) and inducible nitric oxide synthase (iNOS) antibodies. RESULTS Our results showed that montelukast in dose dependant manner improves oxidative stress, lipids profile and TNF α which were affected by EV. Moreover, immunohistochemical examination revealed that montelukast markedly reduced iNOS expression, while expression of PTEN was markedly enhanced, as compared to EV group. The protective effects of montelukast were also verified histopathologically. CONCLUSIONS Montelukast in dose dependant manner provided biochemical and histo-pathological improvement in EV induced EH, through its anti-inflammatory, antioxidant activity and inhibition of iNOS expression with induction of PTEN expression.
Collapse
|
30
|
Vahrenkamp JM, Yang CH, Rodriguez AC, Almomen A, Berrett KC, Trujillo AN, Guillen KP, Welm BE, Jarboe EA, Janat-Amsbury MM, Gertz J. Clinical and Genomic Crosstalk between Glucocorticoid Receptor and Estrogen Receptor α In Endometrial Cancer. Cell Rep 2018; 22:2995-3005. [PMID: 29539426 PMCID: PMC5870123 DOI: 10.1016/j.celrep.2018.02.076] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/31/2018] [Accepted: 02/21/2018] [Indexed: 11/27/2022] Open
Abstract
Steroid hormone receptors are simultaneously active in many tissues and are capable of altering each other's function. Estrogen receptor α (ER) and glucocorticoid receptor (GR) are expressed in the uterus, and their ligands have opposing effects on uterine growth. In endometrial tumors with high ER expression, we surprisingly found that expression of GR is associated with poor prognosis. Dexamethasone reduced normal uterine growth in vivo; however, this growth inhibition was abolished in estrogen-induced endometrial hyperplasia. We observed low genomic-binding site overlap when ER and GR are induced with their respective ligands; however, upon simultaneous induction they co-occupy more sites. GR binding is altered significantly by estradiol with GR recruited to ER-bound loci that become more accessible upon estradiol induction. Gene expression responses to co-treatment were more similar to estradiol but with additional regulated genes. Our results suggest phenotypic and molecular interplay between ER and GR in endometrial cancer.
Collapse
Affiliation(s)
- Jeffery M Vahrenkamp
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Chieh-Hsiang Yang
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT 84112, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Adriana C Rodriguez
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Aliyah Almomen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT 84112, USA; College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Kristofer C Berrett
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Katrin P Guillen
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Bryan E Welm
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Surgery, University of Utah, Salt Lake City, UT 84112, USA
| | - Elke A Jarboe
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Margit M Janat-Amsbury
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT 84112, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Jason Gertz
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
31
|
Jia N, Wang J, Li Q, Tao X, Chang K, Hua K, Yu Y, Wong KK, Feng W. DNA methylation promotes paired box 2 expression via myeloid zinc finger 1 in endometrial cancer. Oncotarget 2018; 7:84785-84797. [PMID: 27764784 PMCID: PMC5356698 DOI: 10.18632/oncotarget.12626] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/25/2016] [Indexed: 12/27/2022] Open
Abstract
This work investigated the role of paired box 2 (PAX2) in endometrial cancer and its epigenetic regulation mechanism. Endometrial cancer tissues and cell lines exhibited increased PAX2 expression compared with hyperplasia, normal endometrium and endometrial epithelial cells. Knock-down of PAX2 resulted in reduced cell viability, invasion and migration, and PAX2 overexpression caused the opposite effects. Increased methylation of the PAX2 promoter was observed in both cancer tissues and cell lines and was positively correlated with PAX2 expression. After 5-Aza-CdR treatment, PAX2 mRNA and protein were down-regulated, and PAX2 methylation was decreased. Deletion analysis confirmed that a repressive transcriptional regulatory region of the PAX2 promoter coincided with the hypermethylated region identified in MassARRAY analysis. Binding sites of myeloid zinc finger 1 (MZF1) are predicted in the defined region. Knock-down of MZF1 up-regulated the transcriptional activity and protein level of PAX2 after 5-Aza-CdR treatment, which indicated that MZF1 may act as a repressive transcription factor when the PAX2 promoter is unmethylated. In conclusion, PAX2 is involved in the carcinogenesis of endometrial cancer by stimulating cell growth and promoting cell motility. The overexpression of PAX2 in endometrial cancer is regulated by promoter hypermethylation and the transcription factor MZF1.
Collapse
Affiliation(s)
- Nan Jia
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Jieyu Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Qing Li
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Xiang Tao
- Department of Pathology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Kaikai Chang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Keqin Hua
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Yinhua Yu
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Kwong-Kwok Wong
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Weiwei Feng
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Cheng Y, Lv Q, Xie B, Yang B, Shan W, Ning C, Li B, Xie L, Gu C, Luo X, Chen X, Zhu Q. Estrogen and high-fat diet induced alterations in C57BL/6 mice endometrial transcriptome profile. Endocr Connect 2018; 7:36-46. [PMID: 29133384 PMCID: PMC5744625 DOI: 10.1530/ec-17-0315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 12/31/2022]
Abstract
Unopposed estrogen stimulation and insulin resistance are known to play important roles in endometrial cancer (EC), but the interaction between these two factors and how they contribute to endometrial lesions are not completely elucidated. To investigate the endometrial transcriptome profile and the associated molecular pathway alterations, we established an ovariectomized C57BL/6 mouse model treated with subcutaneous implantation of 17-β estradiol (E2) pellet and/or high-fat diet (HFD) for 12 weeks to mimic sustained estrogen stimulation and insulin resistance. Histomorphologically, we found that both E2 and E2 + HFD groups showed markedly enlarged uterus and increased number of endometrial glands. The endometrium samples were collected for microarray assay. GO and KEGG analysis showed that genes regulated by E2 and/or HFD are mainly responsible for immune response, inflammatory response and metabolic pathways. Further IPA analysis demonstrated that the acute phase response signaling, NF-κB signaling, leukocyte extravasation signaling, PPAR signaling and LXR/RXR activation pathways are mainly involved in the pathways above. In addition, the genes modulated reciprocally by E2 and/or HFD were also analyzed, and their crosstalk mainly focuses on enhancing one another's activity. The combination analysis of microarray data and TCGA database provided potential diagnostic or therapeutic targets for EC. Further validation was performed in mice endometrium and human EC cell lines. In conclusion, this study unraveled the endometrial transcriptome profile alterations affected by E2 and/or HFD that may disturb endometrial homeostasis and contribute to the development of endometrial hyperplasia.
Collapse
Affiliation(s)
- Yali Cheng
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qiaoying Lv
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Bingying Xie
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Bingyi Yang
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Weiwei Shan
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chengcheng Ning
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Bing Li
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Liying Xie
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chao Gu
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xuezhen Luo
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaojun Chen
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qin Zhu
- Department of PathologyObstetrics and Gynecology, Hospital of Fudan University, Shanghai, China
| |
Collapse
|
33
|
Refaie MM, El-Hussieny M. The role of interleukin-1b and its antagonist (diacerein) in estradiol benzoate-induced endometrial hyperplasia and atypia in female rats. Fundam Clin Pharmacol 2017; 31:438-446. [DOI: 10.1111/fcp.12285] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/05/2017] [Accepted: 03/13/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Marwa M.M. Refaie
- Department of Pharmacology; Faculty of Medicine; El-Minia University; 61511 El-Minia Egypt
| | - Maram El-Hussieny
- Department of Pathology; Faculty of Medicine; El-Minia University; 61511 El-Minia Egypt
| |
Collapse
|
34
|
Chen C, Wang Y, Wang S, Liu Y, Zhang J, Xu Y, Zhang Z, Bao W, Wu S. LSD1 sustains estrogen-driven endometrial carcinoma cell proliferation through the PI3K/AKT pathway via di-demethylating H3K9 of cyclin D1. Int J Oncol 2017; 50:942-952. [PMID: 28098854 DOI: 10.3892/ijo.2017.3849] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/19/2016] [Indexed: 11/05/2022] Open
Abstract
A recent study reported that histone lysine specific demethylase 1 (LSD1, KDM1A) is overexpressed in endometrioid endometrial carcinoma (EEC) and associated with tumor progression as well as poor prognosis. However, the physiological function and mechanism of LSD1 in endometrial cancer (EC) remains largely unknown. In this study, we demonstrate that β-estradiol (E2) treatment increased LSD1 expression via the GPR30/PI3K/AKT pathway in endometrial cancer cells. Both siGPR30 and the PI3K inhibitor LY294002 block this effect. RNAi-mediated silencing of LSD1 abolished estrogen-driven endometrial cancer cell (ECC) proliferation, and induced G1 cell arrest and apoptosis. Mechanistically, we find that LSD1 silencing results in PI3K/AKT signal inactivation, but without the elevation of PTEN expression as expected. This is because the inhibition of LSD1 induces dimethylation of lysine 9 on histone H3 (H3K9m2) accumulation at the promoter region of cyclin D1. Interfering with cyclin D1 leads to PI3K/AKT signal suppression. Re-overexpression of cyclin D1 in LSD1-knockdown ECCs reverses the LSD1 inhibitory action. Our finding connects estrogen signaling with epigenetic regulation in EEC and provides novel experimental support for LSD1 as a potential target for endometrial cancer therapeutics.
Collapse
Affiliation(s)
- Chunqin Chen
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yanan Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Shiyu Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yuan Liu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Shanghai Tongji University, Shanghai, P.R. China
| | - Yuyao Xu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Zhenbo Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Sufang Wu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
35
|
Bentov Y, Jurisicova A, Kenigsberg S, Casper RF. What maintains the high intra-follicular estradiol concentration in pre-ovulatory follicles? J Assist Reprod Genet 2015; 33:85-94. [PMID: 26552664 DOI: 10.1007/s10815-015-0612-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 10/28/2015] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The purpose of the study was to establish the mechanism by which the estrogen concentration difference between the follicular fluid and the serum is maintained. METHODS We used dialysis membrane with a pore size of <3 KD to characterize the estrogen-binding capacity of the follicular fluid. We performed PCR, western blot, and ELISA on luteinized granulosa cells to determine if sex hormone-binding globulin (SHBG) is produced by granulosa cells, and finally we used affinity columns and mass spectrometry to identify the estrogen-binding protein in the follicular fluid. RESULTS We found that a significant estrogen concentration difference is maintained in a cell-free system and is lost with proteolysis of the follicular fluid proteins. Luteinized granulosa cells are likely not a source of SHBG, as we were not able to detect expression of SHBG in these cells. Perlecan was the most highly enriched follicular fluid protein in the affinity columns. CONCLUSIONS We were able to identify perlecan as the most likely candidate for the major estrogen-binding protein in the follicular fluid.
Collapse
Affiliation(s)
- Yaakov Bentov
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada. .,Toronto Center for Assisted Reproductive Technologies, Toronto, Canada. .,Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada.
| | - Andrea Jurisicova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| | - Shlomit Kenigsberg
- CReATe Fertility Centre, Toronto, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| | - Robert F Casper
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.,Toronto Center for Assisted Reproductive Technologies, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| |
Collapse
|