1
|
Dias Da Silva I, Wuidar V, Zielonka M, Pequeux C. Unraveling the Dynamics of Estrogen and Progesterone Signaling in the Endometrium: An Overview. Cells 2024; 13:1236. [PMID: 39120268 PMCID: PMC11312103 DOI: 10.3390/cells13151236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/25/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
The endometrium is crucial for the perpetuation of human species. It is a complex and dynamic tissue lining the inner wall of the uterus, regulated throughout a woman's life based on estrogen and progesterone fluctuations. During each menstrual cycle, this multicellular tissue undergoes cyclical changes, including regeneration, differentiation in order to allow egg implantation and embryo development, or shedding of the functional layer in the absence of pregnancy. The biology of the endometrium relies on paracrine interactions between epithelial and stromal cells involving complex signaling pathways that are modulated by the variations of estrogen and progesterone levels across the menstrual cycle. Understanding the complexity of estrogen and progesterone receptor signaling will help elucidate the mechanisms underlying normal reproductive physiology and provide fundamental knowledge contributing to a better understanding of the consequences of hormonal imbalances on gynecological conditions and tumorigenesis. In this narrative review, we delve into the physiology of the endometrium, encompassing the complex signaling pathways of estrogen and progesterone.
Collapse
Grants
- J.0165.24, 7.6529.23, J.0153.22, 7.4580.21F, 7.6518.21, J.0131.19 Fund for Scientific Research
- FSR-F-2023-FM, FSR-F-2022-FM, FSR-F-2021-FM, FSR-F-M-19/6761 University of Liège
- 2020, 2021, 2022 Fondation Léon Fredericq
Collapse
Affiliation(s)
| | | | | | - Christel Pequeux
- Tumors and Development, Estrogen-Sensitive Tissues and Cancer Team, GIGA-Cancer, Laboratory of Biology, University of Liège, 4000 Liège, Belgium; (I.D.D.S.); (V.W.); (M.Z.)
| |
Collapse
|
2
|
Abbott DA, Mancini MG, Bolt MJ, Szafran AT, Neugebauer KA, Stossi F, Gorelick DA, Mancini MA. A novel ERβ high throughput microscopy platform for testing endocrine disrupting chemicals. Heliyon 2024; 10:e23119. [PMID: 38169792 PMCID: PMC10758781 DOI: 10.1016/j.heliyon.2023.e23119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
In this study we present an inducible biosensor model for the Estrogen Receptor Beta (ERβ), GFP-ERβ:PRL-HeLa, a single-cell-based high throughput (HT) in vitro assay that allows direct visualization and measurement of GFP-tagged ERβ binding to ER-specific DNA response elements (EREs), ERβ-induced chromatin remodeling, and monitor transcriptional alterations via mRNA fluorescence in situ hybridization for a prolactin (PRL)-dsRED2 reporter gene. The model was used to accurately (Z' = 0.58-0.8) differentiate ERβ-selective ligands from ERα ligands when treated with a panel of selective agonists and antagonists. Next, we tested an Environmental Protection Agency (EPA)-provided set of 45 estrogenic reference chemicals with known ERα in vivo activity and identified several that activated ERβ as well, with varying sensitivity, including a subset that is completely novel. We then used an orthogonal ERE-containing transgenic zebrafish (ZF) model to cross validate ERβ and ERα selective activities at the organism level. Using this environmentally relevant ZF assay, some compounds were confirmed to have ERβ activity, validating the GFP-ERβ:PRL-HeLa assay as a screening tool for potential ERβ active endocrine disruptors (EDCs). These data demonstrate the value of sensitive multiplex mechanistic data gathered by the GFP-ERβ:PRL-HeLa assay coupled with an orthogonal zebrafish model to rapidly identify environmentally relevant ERβ EDCs and improve upon currently available screening tools for this understudied nuclear receptor.
Collapse
Affiliation(s)
- Derek A. Abbott
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Maureen G. Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
| | - Michael J. Bolt
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
- Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX, USA
| | - Adam T. Szafran
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
| | - Kaley A. Neugebauer
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
| | - Daniel A. Gorelick
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Michael A. Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
- Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX, USA
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
3
|
Stossi F, Rivera Tostado A, Johnson HL, Mistry RM, Mancini MG, Mancini MA. Gene transcription regulation by ER at the single cell and allele level. Steroids 2023; 200:109313. [PMID: 37758052 PMCID: PMC10842394 DOI: 10.1016/j.steroids.2023.109313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/12/2023] [Accepted: 09/23/2023] [Indexed: 10/03/2023]
Abstract
In this short review we discuss the current view of how the estrogen receptor (ER), a pivotal member of the nuclear receptor superfamily of transcription factors, regulates gene transcription at the single cell and allele level, focusing on in vitro cell line models. We discuss central topics and new trends in molecular biology including phenotypic heterogeneity, single cell sequencing, nuclear phase separated condensates, single cell imaging, and image analysis methods, with particular focus on the methodologies and results that have been reported in the last few years using microscopy-based techniques. These observations augment the results from biochemical assays that lead to a much more complex and dynamic view of how ER, and arguably most transcription factors, act to regulate gene transcription.
Collapse
Affiliation(s)
- Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States.
| | | | - Hannah L Johnson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States
| | - Ragini M Mistry
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States
| | - Maureen G Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, United States; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
4
|
Bardhi O, Palmer BF, Clegg DJ. The evolutionary impact and influence of oestrogens on adipose tissue structure and function. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220207. [PMID: 37482787 PMCID: PMC10363706 DOI: 10.1098/rstb.2022.0207] [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: 03/02/2023] [Accepted: 06/06/2023] [Indexed: 07/25/2023] Open
Abstract
Oestrogens are sex steroid hormones that have gained prominence over the years owing to their crucial roles in human health and reproduction functions which have been preserved throughout evolution. One of oestrogens actions, and the focus of this review, is their ability to determine adipose tissue distribution, function and adipose tissue 'health'. Body fat distribution is sexually dimorphic, affecting males and females differently. These differences are also apparent in the development of the metabolic syndrome and other chronic conditions where oestrogens are critical. In this review, we summarize the different molecular mechanisms, pathways and resulting pathophysiology which are a result of oestrogens actions in and on adipose tissues. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Olgert Bardhi
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Biff F. Palmer
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Deborah J. Clegg
- Vice President for Research, Texas Tech Health Sciences Center, El Paso, TX 75390, USA
| |
Collapse
|
5
|
Zaghloul E, Handousa H, Singab ANB, Elmazar MM, Ayoub IM, Swilam N. Phytoecdysteroids and Anabolic Effect of Atriplex dimorphostegia: UPLC-PDA-MS/MS Profiling, In Silico and In Vivo Models. PLANTS (BASEL, SWITZERLAND) 2023; 12:206. [PMID: 36616335 PMCID: PMC9824417 DOI: 10.3390/plants12010206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Atriplex dimorphostegia (Saltbush) is an annual halophytic shrub that is widely distributed across various parts of Asia. The current study is the first to report the metabolites profile of the total ethanol extract of the aerial parts of A. dimorphostegia (TEAD), and its anabolic activity together with the isolated 20-hydroxyecdysone (20-HE) in orchidectomized male rats. TEAD was analyzed and standardized utilizing UPLC-PDA-ESI−MS/MS and UPLC-PDA-UV techniques, resulting in tentative identification of fifty compounds including polyphenols, steroids and triterpenoids. In addition, 20-HE was quantified, representing 26.79 μg/mg of the extract. Phytochemical investigation of TEAD resulted in the isolation of 20-HE from the ethyl acetate fraction (EFAD) and was identified by conventional spectroscopic methods of analysis. Furthermore, the anabolic effect of the isolated 20-HE and TEAD was then evaluated using in silico and in vivo models. Molecular docking experiments revealed in vitro selectivity of 20-HE towards estrogen receptors (ERs), specifically ERβ over ERα and androgenic receptor (AR). The anabolic efficacy of TEAD and 20-HE was studied in orchidectomized immature male Wistar rats using the weight of gastrocnemius and soleus muscles. The weights of ventral prostate and seminal vesicles were used as indicators for androgenic activity. Rats administered 20-HE and TEAD showed a significant increase (p = 0.0006 and p < 0.0001) in the net muscle mass compared to the negative control, while the group receiving TEAD showed the highest percentage among all groups at p < 0.0001. Histopathological investigation of skeletal muscle fibers showed normal morphological structures, and the group administered 20-HE showed an increase in cross sectional area of muscle fibers comparable to methandienone and testosterone groups at p > 0.99. A. dimorphostegia exhibited promising anabolic activity with minimal androgenic side effects.
Collapse
Affiliation(s)
- Eman Zaghloul
- Department of Pharmacognosy, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo 11837, Egypt
| | - Heba Handousa
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11435, Egypt
| | - Abdel Nasser B. Singab
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
- Center for Drug Discovery Research and Development, Ain Shams University, Cairo 11566, Egypt
| | - Mohey M. Elmazar
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo 11837, Egypt
| | - Iriny M. Ayoub
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Noha Swilam
- Department of Pharmacognosy, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo 11837, Egypt
| |
Collapse
|
6
|
Horii K, Sawamura T, Onishi A, Yuki N, Naitou K, Shiina T, Shimizu Y. Contribution of sex hormones to the sexually dimorphic response of colorectal motility to noxious stimuli in rats. Am J Physiol Gastrointest Liver Physiol 2022; 323:G1-G8. [PMID: 35438007 DOI: 10.1152/ajpgi.00033.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Our recent studies have shown that noxious stimuli in the colorectum enhance colorectal motility via the brain and spinal defecation centers in male rats. In female rats, however, noxious stimuli have no effect on colorectal motility. The purpose of this study was to determine whether sex hormones are major contributing factors for sex-dependent differences in neural components of the spinal defecation center. Colorectal motility was measured using an in vivo method under ketamine and α-chloralose anesthesia in rats. Capsaicin was administered into the colorectal lumen as noxious stimuli. Orchiectomy in male rats had no effect on the capsaicin-induced response of colorectal motility. However, in ovariectomized female rats, capsaicin administration enhanced colorectal motility, though intact female animals did not show enhanced motility. When estradiol was administered by using a sustained-release preparation in ovariectomized female rats, capsaicin administration did not enhance colorectal motility unless a GABAA receptor antagonist was intrathecally administered to the lumbosacral spinal cord. These findings suggest that estradiol allowed the GABAergic neurons to operate in response to intracolonic administration of capsaicin. The operation of GABAergic inhibition by the action of estradiol could be manifested in male rats only when the effects of male sex hormones were removed by orchiectomy. Taken together, our results indicate that sex hormones contribute to the sexually dimorphic response in colorectal motility enhancement in response to noxious stimuli through modulating GABAergic pathways.NEW & NOTEWORTHY This study demonstrated that estradiol permits inhibitory regulation in the spinal defecation center not only in female rats but also in orchiectomized male rats. GABAergic pathways are likely involved in the effect of estradiol. This is the first report showing that sex hormones affect colorectal motility through the alteration of neural components of the regulatory pathways. Our findings provide a novel insight into pathophysiological mechanisms of defecation disorders related to changes in sex hormones.
Collapse
Affiliation(s)
- Kazuhiro Horii
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Tomoya Sawamura
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Ayaka Onishi
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Natsufu Yuki
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Kiyotada Naitou
- Department of Basic Veterinary Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Takahiko Shiina
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan.,Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Yasutake Shimizu
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan.,Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
7
|
Larson AA, Shams AS, McMillin SL, Sullivan BP, Vue C, Roloff ZA, Batchelor E, Kyba M, Lowe DA. Estradiol deficiency reduces the satellite cell pool by impairing cell cycle progression. Am J Physiol Cell Physiol 2022; 322:C1123-C1137. [PMID: 35442828 PMCID: PMC9169829 DOI: 10.1152/ajpcell.00429.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/31/2022] [Accepted: 04/17/2022] [Indexed: 12/22/2022]
Abstract
The size of the satellite cell pool is reduced in estradiol (E2)-deficient female mice and humans. Here, we use a combination of in vivo and in vitro approaches to identify mechanisms, whereby E2 deficiency impairs satellite cell maintenance. By measuring satellite cell numbers in mice at several early time points postovariectomy (Ovx), we determine that satellite cell numbers decline by 33% between 10 and 14 days post-Ovx in tibialis anterior and gastrocnemius muscles. At 14 days post-Ovx, we demonstrate that satellite cells have a reduced propensity to transition from G0/G1 to S and G2/M phases, compared with cells from ovary-intact mice, associated with changes in two key satellite cell cycle regulators, ccna2 and p16INK4a. Further, freshly isolated satellite cells treated with E2 in vitro have 62% greater cell proliferation and require less time to complete the first division. Using clonal and differentiation assays, we measured 69% larger satellite cell colonies and enhanced satellite cell-derived myoblast differentiation with E2 treatment compared with vehicle-treated cells. Together, these results identify a novel mechanism for preservation of the satellite cell pool by E2 via promotion of satellite cell cycling.
Collapse
Affiliation(s)
- Alexie A Larson
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Ahmed S Shams
- Lillehei Heart Institute, Medical School, University of Minnesota, Minneapolis, Minnesota
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, Minnesota
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Shawna L McMillin
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Brian P Sullivan
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Cha Vue
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Zachery A Roloff
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Eric Batchelor
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Michael Kyba
- Lillehei Heart Institute, Medical School, University of Minnesota, Minneapolis, Minnesota
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Dawn A Lowe
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
8
|
Taldaev A, Terekhov R, Nikitin I, Zhevlakova A, Selivanova I. Insights into the Pharmacological Effects of Flavonoids: The Systematic Review of Computer Modeling. Int J Mol Sci 2022; 23:6023. [PMID: 35682702 PMCID: PMC9181432 DOI: 10.3390/ijms23116023] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 12/13/2022] Open
Abstract
Computer modeling is a method that is widely used in scientific investigations to predict the biological activity, toxicity, pharmacokinetics, and synthesis strategy of compounds based on the structure of the molecule. This work is a systematic review of articles performed in accordance with the recommendations of PRISMA and contains information on computer modeling of the interaction of classical flavonoids with different biological targets. The review of used computational approaches is presented. Furthermore, the affinities of flavonoids to different targets that are associated with the infection, cardiovascular, and oncological diseases are discussed. Additionally, the methodology of bias risks in molecular docking research based on principles of evidentiary medicine was suggested and discussed. Based on this data, the most active groups of flavonoids and lead compounds for different targets were determined. It was concluded that flavonoids are a promising object for drug development and further research of pharmacology by in vitro, ex vivo, and in vivo models is required.
Collapse
Affiliation(s)
- Amir Taldaev
- Laboratoty of Nanobiotechnology, Institute of Biomedical Chemistry, Pogodinskaya Str. 10/8, 119121 Moscow, Russia
- Department of Chemistry, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (R.T.); (I.N.); (A.Z.); (I.S.)
| | - Roman Terekhov
- Department of Chemistry, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (R.T.); (I.N.); (A.Z.); (I.S.)
| | - Ilya Nikitin
- Department of Chemistry, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (R.T.); (I.N.); (A.Z.); (I.S.)
| | - Anastasiya Zhevlakova
- Department of Chemistry, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (R.T.); (I.N.); (A.Z.); (I.S.)
| | - Irina Selivanova
- Department of Chemistry, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (R.T.); (I.N.); (A.Z.); (I.S.)
| |
Collapse
|
9
|
Characterization of transcriptome diversity and in vitro behavior of primary human high-risk breast cells. Sci Rep 2022; 12:6159. [PMID: 35459280 PMCID: PMC9033878 DOI: 10.1038/s41598-022-10246-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 04/01/2022] [Indexed: 11/09/2022] Open
Abstract
Biology and transcriptomes of non-cancerous human mammary epithelial cells at risk for breast cancer development were explored following primary isolation utilizing conditional reprogramming cell technology from mastectomy tissue ipsilateral to invasive breast cancer. Cultures demonstrated consistent categorizable behaviors. Relative viability and mammosphere formation differed between samples but were stable across three different mammary-specific media. E2F cell cycle target genes expression levels were positively correlated with viability and advancing age was inversely associated. Estrogen growth response was associated with Tissue necrosis factor signaling and Interferon alpha response gene enrichment. Neoadjuvant chemotherapy exposure significantly altered transcriptomes, shifting them towards expression of genes linked to mammary stem cell formation. Breast cancer prognostic signature sets include genes that in normal development are limited to specific stages of pregnancy or the menstrual cycle. Sample transcriptomes were queried for stage specific gene expression patterns. All cancer samples and a portion of high-risk samples showed overlapping stages reflective of abnormal gene expression patterns, while other high-risk samples exhibited more stage specific patterns. In conclusion, at-risk cells preserve behavioral and transcriptome diversity that could reflect different risk profiles. It is possible that prognostic platforms analogous to those used for breast cancer could be developed for high-risk mammary cells.
Collapse
|
10
|
Factors Influencing Gallstone Formation: A Review of the Literature. Biomolecules 2022; 12:biom12040550. [PMID: 35454138 PMCID: PMC9026518 DOI: 10.3390/biom12040550] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/04/2023] Open
Abstract
Gallstone disease is a common pathology of the digestive system with nearly a 10–20% incidence rate among adults. The mainstay of treatment is cholecystectomy, which is commonly associated with physical pain and may also seriously affect a patient’s quality of life. Clinical research suggests that cholelithiasis is closely related to the age, gender, body mass index, and other basic physical characteristics of patients. Clinical research further suggests that the occurrence of cholelithiasis is related to obesity, diabetes, non-alcoholic fatty liver, and other diseases. For this reason, we reviewed the following: genetic factors; excessive liver cholesterol secretion (causing cholesterol supersaturation in gallbladder bile); accelerated growth of cholesterol crystals and solid cholesterol crystals; gallbladder motility impairment; and cardiovascular factors. Herein, we summarize and analyze the causes and mechanisms of cholelithiasis, discuss its correlation with the pathogenesis of related diseases, and discuss possible mechanisms.
Collapse
|
11
|
Lewandowski CT, Laham MS, Thatcher GR. Remembering your A, B, C's: Alzheimer's disease and ABCA1. Acta Pharm Sin B 2022; 12:995-1018. [PMID: 35530134 PMCID: PMC9072248 DOI: 10.1016/j.apsb.2022.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 12/24/2022] Open
Abstract
The function of ATP binding cassette protein A1 (ABCA1) is central to cholesterol mobilization. Reduced ABCA1 expression or activity is implicated in Alzheimer's disease (AD) and other disorders. Therapeutic approaches to boost ABCA1 activity have yet to be translated successfully to the clinic. The risk factors for AD development and progression, including comorbid disorders such as type 2 diabetes and cardiovascular disease, highlight the intersection of cholesterol transport and inflammation. Upregulation of ABCA1 can positively impact APOE lipidation, insulin sensitivity, peripheral vascular and blood–brain barrier integrity, and anti-inflammatory signaling. Various strategies towards ABCA1-boosting compounds have been described, with a bias toward nuclear hormone receptor (NHR) agonists. These agonists display beneficial preclinical effects; however, important side effects have limited development. In particular, ligands that bind liver X receptor (LXR), the primary NHR that controls ABCA1 expression, have shown positive effects in AD mouse models; however, lipogenesis and unwanted increases in triglyceride production are often observed. The longstanding approach, focusing on LXRβ vs. LXRα selectivity, is over-simplistic and has failed. Novel approaches such as phenotypic screening may lead to small molecule NHR modulators that elevate ABCA1 function without inducing lipogenesis and are clinically translatable.
Collapse
|
12
|
Tan SC, Low TY, Mohamad Hanif EA, Sharzehan MAK, Kord-Varkaneh H, Islam MA. The rs9340799 polymorphism of the estrogen receptor alpha (ESR1) gene and its association with breast cancer susceptibility. Sci Rep 2021; 11:18619. [PMID: 34545128 PMCID: PMC8452701 DOI: 10.1038/s41598-021-97935-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023] Open
Abstract
The ESR1 rs9340799 polymorphism has been frequently investigated with regard to its association with breast cancer (BC) susceptibility, but the findings have been inconclusive. In this work, we aimed to address the inconsistencies in study findings by performing a systematic review and meta-analysis. Eligible studies were identified from the Web of Science, PubMed, Scopus, China National Knowledge Infrastructure, VIP and Wanfang databases based on the predefined inclusion and exclusion criteria. The pooled odds ratio (OR) was then calculated under five genetic models: homozygous (GG vs. AA), heterozygous (AG vs. AA), dominant (AG + GG vs. AA), recessive (GG vs. AA + AG) and allele (G vs. A). Combined results from 23 studies involving 34,721 subjects indicated a lack of significant association between the polymorphism and BC susceptibility (homozygous model, OR = 1.045, 95% CI 0.887-1.231, P = 0.601; heterozygous model, OR = 0.941, 95% CI 0.861-1.030, P = 0.186; dominant model, OR = 0.957, 95% CI 0.875-1.045, P = 0.327; recessive model, OR = 1.053, 95% CI 0.908-1.222, P = 0.495; allele model, OR = 0.987, 95% CI 0.919-1.059, P = 0.709). Subgroup analyses by ethnicity, menopausal status and study quality also revealed no statistically significant association (P > 0.05). In conclusion, our results showed that the ESR1 rs9340799 polymorphism was not associated with BC susceptibility, suggesting its limited potential as a genetic marker for BC.
Collapse
Affiliation(s)
- Shing Cheng Tan
- grid.412113.40000 0004 1937 1557UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Teck Yew Low
- grid.412113.40000 0004 1937 1557UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ezanee Azlina Mohamad Hanif
- grid.412113.40000 0004 1937 1557UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohamad Ayub Khan Sharzehan
- grid.412113.40000 0004 1937 1557UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Hamed Kord-Varkaneh
- grid.411600.2Department of Clinical Nutrition and Dietetics, Student Research Committee, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Md Asiful Islam
- grid.11875.3a0000 0001 2294 3534Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
13
|
Das N, Mishra SK, Bishayee A, Ali ES, Bishayee A. The phytochemical, biological, and medicinal attributes of phytoecdysteroids: An updated review. Acta Pharm Sin B 2021; 11:1740-1766. [PMID: 34386319 PMCID: PMC8343124 DOI: 10.1016/j.apsb.2020.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/04/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
The phytoecdysteroids (PEs) comprise a large group of biologically-active plant steroids, which have structures similar to those of insect-molting hormones. PEs are distributed in plants as secondary metabolites that offer protection against phytophagus (plant-eating) insects. When insects consume the plants containing these chemicals, they promptly molt and undergo metabolic destruction; the insects eventually die. Chemically, ecdysteroids are a group of polyhydroxylated ketosteroids that are structurally similar to androgens. The carbon skeleton of ecdysteroids is termed as cyclopentanoperhydro-phenanthrene with a β-side chain at carbon-17. The essential characteristics of ecdysteroids are a cis-(5β-H) junction of rings A and B, a 7-en-6-one chromophore, and a trans-(14α-OH) junction of rings C and D. Plants only synthesize PEs from mevalonic acid in the mevalonate pathway of the plant cell using acetyl-CoA as a precursor; the most common PE is 20-hydroxyecdysone. So far, over 400 PEs have been identified and reported, and a compilation of 166 PEs originating from 1998 has been previously reviewed. In the present review, we have summarized 212 new PEs reported between 1999 and 2019. We have also critically analyzed the biological, pharmacological, and medicinal properties of PEs to understand the full impact of these phytoconstituents in health and disease.
Collapse
Affiliation(s)
- Niranjan Das
- Department of Chemistry, Iswar Chandra Vidyasagar College, Belonia-799 155, Tripura, India
| | - Siddhartha Kumar Mishra
- Cancer Biology Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar-470 003, Madhya Pradesh, India
| | | | - Eunüs S. Ali
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| |
Collapse
|
14
|
Díaz M, Lobo F, Hernández D, Amesty Á, Valdés-Baizabal C, Canerina-Amaro A, Mesa-Herrera F, Soler K, Boto A, Marín R, Estévez-Braun A, Lahoz F. FLTX2: A Novel Tamoxifen Derivative Endowed with Antiestrogenic, Fluorescent, and Photosensitizer Properties. Int J Mol Sci 2021; 22:ijms22105339. [PMID: 34069498 PMCID: PMC8161337 DOI: 10.3390/ijms22105339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/28/2021] [Accepted: 05/08/2021] [Indexed: 11/16/2022] Open
Abstract
Tamoxifen is the most widely used selective modulator of estrogen receptors (SERM) and the first strategy as coadjuvant therapy for the treatment of estrogen-receptor (ER) positive breast cancer worldwide. In spite of such success, tamoxifen is not devoid of undesirable effects, the most life-threatening reported so far affecting uterine tissues. Indeed, tamoxifen treatment is discouraged in women under risk of uterine cancers. Recent molecular design efforts have endeavoured the development of tamoxifen derivatives with antiestrogen properties but lacking agonistic uterine tropism. One of this is FLTX2, formed by the covalent binding of tamoxifen as ER binding core, 7-nitrobenzofurazan (NBD) as the florescent dye, and Rose Bengal (RB) as source for reactive oxygen species. Our analyses demonstrate (1) FLTX2 is endowed with similar antiestrogen potency as tamoxifen and its predecessor FLTX1, (2) shows a strong absorption in the blue spectral range, associated to the NBD moiety, which efficiently transfers the excitation energy to RB through intramolecular FRET mechanism, (3) generates superoxide anions in a concentration- and irradiation time-dependent process, and (4) Induces concentration- and time-dependent MCF7 apoptotic cell death. These properties make FLTX2 a very promising candidate to lead a novel generation of SERMs with the endogenous capacity to promote breast tumour cell death in situ by photosensitization.
Collapse
Affiliation(s)
- Mario Díaz
- Departamento Biología Animal, Edafología y Geología, Universidad de La Laguna, 38200 Tenerife, Spain;
- Unidad Asociada ULL-CSIC “Fisiología y Biofísica de la Membrana Celular en Enfermedades Neurodegenerativas y Tumorales”, 38200 Tenerife, Spain; (A.B.); (R.M.); (F.L.)
- Correspondence:
| | - Fernando Lobo
- Programa Agustín de Betancourt, Universidad de la Laguna, 38200 Tenerife, Spain; (F.L.); (Á.A.); (C.V.-B.)
| | - Dácil Hernández
- Instituto de Productos Naturales y Agrobiología del CSIC, Avda. Astrofísico F. Sánchez, 38206 Tenerife, Spain;
| | - Ángel Amesty
- Programa Agustín de Betancourt, Universidad de la Laguna, 38200 Tenerife, Spain; (F.L.); (Á.A.); (C.V.-B.)
- Instituto Universitario de Bioorgánica “Antonio González”, Universidad de La Laguna, 38200 Tenerife, Spain;
| | - Catalina Valdés-Baizabal
- Programa Agustín de Betancourt, Universidad de la Laguna, 38200 Tenerife, Spain; (F.L.); (Á.A.); (C.V.-B.)
- Departamento Ciencias Médicas Básicas, Universidad de La Laguna, 38200 Tenerife, Spain;
| | - Ana Canerina-Amaro
- Departamento Ciencias Médicas Básicas, Universidad de La Laguna, 38200 Tenerife, Spain;
| | - Fátima Mesa-Herrera
- Departamento Biología Animal, Edafología y Geología, Universidad de La Laguna, 38200 Tenerife, Spain;
| | - Kevin Soler
- Departamento Física, IUdEA, Universidad de La Laguna, 38200 Tenerife, Spain;
| | - Alicia Boto
- Unidad Asociada ULL-CSIC “Fisiología y Biofísica de la Membrana Celular en Enfermedades Neurodegenerativas y Tumorales”, 38200 Tenerife, Spain; (A.B.); (R.M.); (F.L.)
- Instituto de Productos Naturales y Agrobiología del CSIC, Avda. Astrofísico F. Sánchez, 38206 Tenerife, Spain;
| | - Raquel Marín
- Unidad Asociada ULL-CSIC “Fisiología y Biofísica de la Membrana Celular en Enfermedades Neurodegenerativas y Tumorales”, 38200 Tenerife, Spain; (A.B.); (R.M.); (F.L.)
- Departamento Ciencias Médicas Básicas, Universidad de La Laguna, 38200 Tenerife, Spain;
| | - Ana Estévez-Braun
- Instituto Universitario de Bioorgánica “Antonio González”, Universidad de La Laguna, 38200 Tenerife, Spain;
- Departamento Química Orgánica, Universidad de La Laguna, 38200 Tenerife, Spain
| | - Fernando Lahoz
- Unidad Asociada ULL-CSIC “Fisiología y Biofísica de la Membrana Celular en Enfermedades Neurodegenerativas y Tumorales”, 38200 Tenerife, Spain; (A.B.); (R.M.); (F.L.)
- Departamento Física, IUdEA, Universidad de La Laguna, 38200 Tenerife, Spain;
| |
Collapse
|
15
|
Uncovering Evidence for Endocrine-Disrupting Chemicals That Elicit Differential Susceptibility through Gene-Environment Interactions. TOXICS 2021; 9:toxics9040077. [PMID: 33917455 PMCID: PMC8067468 DOI: 10.3390/toxics9040077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/27/2021] [Accepted: 04/02/2021] [Indexed: 12/17/2022]
Abstract
Exposure to endocrine-disrupting chemicals (EDCs) is linked to myriad disorders, characterized by the disruption of the complex endocrine signaling pathways that govern development, physiology, and even behavior across the entire body. The mechanisms of endocrine disruption involve a complex system of pathways that communicate across the body to stimulate specific receptors that bind DNA and regulate the expression of a suite of genes. These mechanisms, including gene regulation, DNA binding, and protein binding, can be tied to differences in individual susceptibility across a genetically diverse population. In this review, we posit that EDCs causing such differential responses may be identified by looking for a signal of population variability after exposure. We begin by summarizing how the biology of EDCs has implications for genetically diverse populations. We then describe how gene-environment interactions (GxE) across the complex pathways of endocrine signaling could lead to differences in susceptibility. We survey examples in the literature of individual susceptibility differences to EDCs, pointing to a need for research in this area, especially regarding the exceedingly complex thyroid pathway. Following a discussion of experimental designs to better identify and study GxE across EDCs, we present a case study of a high-throughput screening signal of putative GxE within known endocrine disruptors. We conclude with a call for further, deeper analysis of the EDCs, particularly the thyroid disruptors, to identify if these chemicals participate in GxE leading to differences in susceptibility.
Collapse
|
16
|
Hu Z, Liu J, Cao J, Zhang H, Liu X. Ovarian transcriptomic analysis of black Muscovy duck at the early, peak and late egg-laying stages. Gene 2021; 777:145449. [PMID: 33482277 DOI: 10.1016/j.gene.2021.145449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/07/2021] [Accepted: 01/15/2021] [Indexed: 11/16/2022]
Abstract
Ovarian development is a complex process involving many genes and pathways. A well-developed ovary is essential for poultry to keep high egg production and egg fertility. In order to better understand the mechanism of egg production performance, a comparative transcriptomic analysis was performed on ovaries of black Muscovy ducks at the early (BE), peak (BP) and late laying (BL) stages. 1683 DEGs were identified from BL-vs-BE, BL-vs-BP and BP-vs-BE, and the up-regulated genes were 41, 835, 260, the down-regulated genes were 60, 255, 730, respectively. Besides, there were 32, 20 and 424 DEGs co-expressed in the two comparison groups, and 11 DEGs were co-expressed in the three comparison groups. HOXA10, HtrA3, StAR, ZP2 and TAT were found to be involved in the regulation of ovarian development were significantly differentially expressed at different laying stages, which helped to regulate ovarian maturation and egg production. Moreover, we discovered several important functional pathways, such as steroid hormone biosynthesis and ovarian steroidogenesis, that appear to be much more active in the BP ovary compared to those of the BE and BL. Furthermore, 17 coding and 244 non-coding new transcripts were detected in the three comparison groups, the gene structures were optimized and the gene annotation informations were improved. These findings will provide a solid foundation on ovarian development in black Muscovy ducks and other poultry animals at different laying stages, and help to understand the complex molecular and cellular mechanisms of ovary.
Collapse
Affiliation(s)
- Zhigang Hu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Jing Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Junting Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Huilin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Xiaolin Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China.
| |
Collapse
|
17
|
Wang HH, de Bari O, Arnatt CK, Liu M, Portincasa P, Wang DQH. Activation of Estrogen Receptor G Protein-Coupled Receptor 30 Enhances Cholesterol Cholelithogenesis in Female Mice. Hepatology 2020; 72:2077-2089. [PMID: 32112420 PMCID: PMC8157628 DOI: 10.1002/hep.31212] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/21/2020] [Accepted: 02/13/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND AIMS Estrogen is an important risk factor for cholesterol gallstone disease because women are twice as likely as men to form gallstones. The classical estrogen receptor α (ERα), but not ERβ, in the liver plays a critical role in the formation of estrogen-induced gallstones in female mice. The molecular mechanisms underlying the lithogenic effect of estrogen on gallstone formation have become more complicated with the identification of G protein-coupled receptor 30 (GPR30), an estrogen receptor. APPROACH AND RESULTS We investigated the biliary and gallstone phenotypes in ovariectomized female GPR30-/- , ERα-/- , and wild-type mice injected intramuscularly with the potent GPR30-selective agonist G-1 at 0 or 1 μg/day and fed a lithogenic diet for 8 weeks. The activation of GPR30 by G-1 enhanced cholelithogenesis by suppressing expression of cholesterol 7α-hydroxylase, the rate-limiting enzyme for the classical pathway of bile salt synthesis. These metabolic abnormalities led to an increase in biliary cholesterol concentrations in company with hepatic hyposecretion of biliary bile salts, thereby inducing cholesterol-supersaturated gallbladder bile and accelerating cholesterol crystallization. G-1 also impairs gallbladder emptying, leading to sluggish gallbladder motility and promoting the development of biliary sludge in the early stage of gallstone formation. The prevalence rates of gallstones were 80% in wild-type and ERα-/- mice treated with G-1 compared to 10% in wild-type mice receiving no G-1. However, no gallstones were formed in GPR30-/- mice treated with G-1. CONCLUSIONS GPR30 produces additional lithogenic actions, working independently of ERα, to increase susceptible to gallstone formation in female mice; both GPR30 and ERα are potential therapeutic targets for cholesterol gallstone disease, particularly in women and patients exposed to high levels of estrogen.
Collapse
Affiliation(s)
- Helen H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY;,Department of Internal Medicine, Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, MO
| | - Ornella de Bari
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, MO
| | | | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, Clinica Medica “A. Murri”, University of Bari Medical School, Bari, Italy
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY;,Department of Internal Medicine, Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, MO
| |
Collapse
|
18
|
Sprouse J, Sampath C, Gangula PR. Role of sex hormones and their receptors on gastric Nrf2 and neuronal nitric oxide synthase function in an experimental hyperglycemia model. BMC Gastroenterol 2020; 20:313. [PMID: 32967621 PMCID: PMC7513483 DOI: 10.1186/s12876-020-01453-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 09/15/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastroparesis, a condition of abnormal gastric emptying, is most commonly observed in diabetic women. To date, the role of ovarian hormones and/or gastric hormone receptors on regulating nitrergic-mediated gastric motility remains inconclusive. AIM The purpose of this study is to investigate whether sex hormones/their receptors can attenuate altered Nuclear factor (erythroid-derived 2)-like 2 (Nrf2), neuronal Nitric Oxide Synthase (nNOS) expression and nitrergic relaxation in gastric neuromuscular tissues exposed to in-vitro hyperglycemia (HG). METHODS Gastric neuromuscular sections from adult female C57BL/6 J mice were incubated in normoglycemic (NG, 5 mM) or hyperglycemic (30 mM or 50 mM) conditions in the presence or absence of selective estrogen receptor (ER) agonists (ERα /PPT or ERβ: DPN); or non-selective sex hormone receptor antagonists (ER/ICI 182,780, or progesterone receptor (PR)/ RU486) for 48 h. mRNA, protein expression and nitrergic relaxation of circular gastric neuromuscular strips were assessed. RESULTS Our findings in HG, compared to NG, demonstrate a significant reduction in ER, Nrf2, and nNOS expression in gastric specimens. In addition, in-vitro treatment with sex hormones and/or their agonists significantly (*p < 0.05) restored Nrf2/nNOSα expression and total nitrite production. Conversely, ER, but not PR, antagonist significantly reduced Nrf2/nNOSα expression and nitrergic relaxation. CONCLUSIONS Our data suggest that ER's can regulate nitrergic function by improving Nrf2/nNOS expression in experimental hyperglycemia.
Collapse
Affiliation(s)
- Jeremy Sprouse
- School of Graduate Studies, Meharry Medical College, Nashville, TN, 37208, USA.,Department of ODS & Research, School of Dentistry, Nashville, TN, 37208, USA
| | - Chethan Sampath
- Department of ODS & Research, School of Dentistry, Nashville, TN, 37208, USA
| | - Pandu R Gangula
- Department of ODS & Research, School of Dentistry, Nashville, TN, 37208, USA.
| |
Collapse
|
19
|
Böckers M, Paul NW, Efferth T. Indeno[1,2,3-cd]pyrene and picene mediate actions via estrogen receptor α signaling pathway in in vitro cell systems, altering gene expression. Toxicol Appl Pharmacol 2020; 396:114995. [DOI: 10.1016/j.taap.2020.114995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 12/26/2022]
|
20
|
Huang CM, Huang CS, Hsu TN, Huang MS, Fong IH, Lee WH, Liu SC. Disruption of Cancer Metabolic SREBP1/miR-142-5p Suppresses Epithelial-Mesenchymal Transition and Stemness in Esophageal Carcinoma. Cells 2019; 9:cells9010007. [PMID: 31861383 PMCID: PMC7016574 DOI: 10.3390/cells9010007] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 12/24/2022] Open
Abstract
: Elevated activity of sterol regulatory element-binding protein 1 (SREBP1) has been implicated in the tumorigenesis of different cancer types. However, the functional roles of SREBP1 in esophageal cancer are not well appreciated. Here, we aimed to investigate the therapeutic potential of SREBP1 and associated signaling in esophageal cancer. Our initial bioinformatics analyses showed that SREBP1 expression was overexpressed in esophageal tumors and correlated with a significantly lower overall survival rate in patients. Additionally, tumor suppressor miR-142-5p was predicted to target SREBP1/ZEB1 and a lower miR-142-5p was correlated with poor prognosis. We then performed in vitro experiments and showed that overexpressing SREBP1 in OE33 cell line led to increased abilities of colony formation, migration, and invasion; the opposite was observed in SREBP1-silenced OE21cells and SREBP1-silencing was accompanied by the reduced mesenchymal markers, including vimentin (Vim) and ZEB1, while E-cadherin and tumor suppressor miR-142-5p were increased. Subsequently, we first demonstrated that both SREBP1 and ZEB1 were potential targets of miR-142-5p, followed by the examination of the regulatory circuit of miR-142-5p and SREBP1/ZEB1. We observed that increased miR-142-5p level led to the reduced tumorigenic properties, such as migration and tumor sphere formation, and both observations were accompanied by the reduction of ZEB1 and SREBP1, and increase of E-cadherin. We then explored the potential therapeutic agent targeting SREBP1-associated signaling by testing fatostatin (4-hydroxytamoxifen, an active metabolite of tamoxifen). We found that fatostatin suppressed the cell viability of OE21 and OE33 cells and tumor spheres. Interestingly, fatostatin treatment reduced CD133+ population in both OE21 and OE33 cells in concert of increased miR-142-5p level. Finally, we evaluated the efficacy of fatostatin using a xenograft mouse model. Mice treated with fatostatin showed a significantly lower tumor burden and better survival rate as compared to their control counterparts. The treatment of fatostatin resulted in the reduced staining of SREBP1, ZEB1, and Vim, while E-cadherin and miR-142-5p were increased. In summary, we showed that increased SREBP1 and reduced miR-142-5p were associated with increased tumorigenic properties of esophageal cancer cells and poor prognosis. Preclinical tests showed that suppression of SREBP1 using fatostatin led to the reduced malignant phenotype of esophageal cancer via the reduction of EMT markers and increased tumor suppressor, miR-142-5p. Further investigation is warranted for the clinical use of fatostatin for the treatment of esophageal malignancy.
Collapse
Affiliation(s)
- Chih-Ming Huang
- Department of Otolaryngology, Taitung Mackay Memorial Hospital, Taitung City 950, Taiwan;
| | - Chin-Sheng Huang
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-S.H.); (T.-N.H.); (M.-S.H.)
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Tung-Nien Hsu
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-S.H.); (T.-N.H.); (M.-S.H.)
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Mao-Suan Huang
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan; (C.-S.H.); (T.-N.H.); (M.-S.H.)
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Iat-Hang Fong
- Department of Medical Research & Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan;
- Department of Pathology, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Wei-Hwa Lee
- Department of Medical Research & Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan;
- Department of Pathology, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan
- Correspondence: (W.-H.L.); (S.-C.L.); Tel.: +886-2-2490088 (ext. 8742) (W.-H.L.); +886-2-87927192 (S.-C.L.); Fax: +886-2-2248-0900 (W.-H.L.); +886-2-87927193 (S.-C.L.)
| | - Shao-Cheng Liu
- Department of Otolaryngology—Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan
- Correspondence: (W.-H.L.); (S.-C.L.); Tel.: +886-2-2490088 (ext. 8742) (W.-H.L.); +886-2-87927192 (S.-C.L.); Fax: +886-2-2248-0900 (W.-H.L.); +886-2-87927193 (S.-C.L.)
| |
Collapse
|
21
|
Sun SX, Zhang YN, Lu DL, Wang WL, Limbu SM, Chen LQ, Zhang ML, Du ZY. Concentration-dependent effects of 17β-estradiol and bisphenol A on lipid deposition, inflammation and antioxidant response in male zebrafish (Danio rerio). CHEMOSPHERE 2019; 237:124422. [PMID: 31352104 DOI: 10.1016/j.chemosphere.2019.124422] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 06/10/2023]
Abstract
Environmental estrogenic compounds are important pollutants, which are widely distributed in natural water bodies. They produce various adverse effects on fish, but their concentration-dependent toxicities in fish metabolism and health are not fully understood. This study investigated the effects of 17β-estradiol (E2) and bisphenol A (BPA) at low and high concentrations on lipid deposition, inflammation and antioxidant response in male zebrafish. We measured fish growth parameters, gonad development, lipid contents and the activities of inflammatory and antioxidant enzymes, as well as their mRNA expressions. All E2 and BPA concentrations used increased body weight, damaged gonad structure and induced feminization in male zebrafish. The exposure of zebrafish to E2 and BPA promoted lipid accumulation by increasing total fat, liver triglycerides and free fatty acid contents, and also upregulated lipogenic genes expression, although they decreased total cholesterol content. Notably, zebrafish exposed to low concentrations of E2 (200 ng/L) and BPA (100 μg/L) had higher lipid synthesis and deposition compared to high concentrations (2000 ng/L and 2000 μg/L, respectively). However, the high concentrations of E2 and BPA increased inflammation and antioxidant response. Furthermore, BPA caused greater damage to fish gonad development and more severe lipid peroxidation compared to E2. Overall, the results suggest that the toxic effects of E2 and BPA on zebrafish are concentration-dependent such that, the relative low concentrations used induced lipid deposition, whereas the high ones caused adverse effects on inflammation and antioxidant response.
Collapse
Affiliation(s)
- Sheng-Xiang Sun
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yun-Ni Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Dong-Liang Lu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wei-Li Wang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Samwel Mchele Limbu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China; Department of Aquatic Sciences and Fisheries Technology, University of Dar as Salaam, Dar es Salaam, Tanzania
| | - Li-Qiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mei-Ling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
22
|
Wang W, Hao X, Han L, Yan Z, Shen WJ, Dong D, Hasbargen K, Bittner S, Cortez Y, Greenberg AS, Azhar S, Kraemer FB. Tissue-Specific Ablation of ACSL4 Results in Disturbed Steroidogenesis. Endocrinology 2019; 160:2517-2528. [PMID: 31504388 PMCID: PMC6773434 DOI: 10.1210/en.2019-00464] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/21/2019] [Indexed: 01/14/2023]
Abstract
ACSL4 is a member of the ACSL family that catalyzes the conversion of long-chain fatty acids to acyl-coenzyme As, which are essential for fatty-acid incorporation and utilization in diverse metabolic pathways, including cholesteryl ester synthesis. Steroidogenic tissues such as the adrenal gland are particularly enriched in cholesteryl esters of long-chain polyunsaturated fatty acids, which constitute an important pool supplying cholesterol for steroid synthesis. The current studies addressed whether ACSL4 is required for normal steroidogenesis. CYP11A1 promoter‒mediated Cre was used to generate steroid tissue‒specific ACSL4 knockout (KO) mice. Results demonstrated that ACSL4 plays an important role in adrenal cholesteryl ester formation, as well as in determining the fatty acyl composition of adrenal cholesteryl esters, with ACSL4 deficiency leading to reductions in cholesteryl ester storage and alterations in cholesteryl ester composition. Statistically significant reductions in corticosterone and testosterone production, but not progesterone production, were observed in vivo, and these deficits were accentuated in ex vivo and in vitro studies of isolated steroid tissues and cells from ACSL4-deficient mice. However, these effects on steroid production appear to be due to reductions in cholesteryl ester stores rather than disturbances in signaling pathways. We conclude that ACSL4 is dispensable for normal steroidogenesis.
Collapse
Affiliation(s)
- Wei Wang
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Xiao Hao
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Lina Han
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Zhe Yan
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Wen-Jun Shen
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Correspondence: Fredric B. Kraemer, MD, or Wen-Jun Shen, PhD, Division of Endocrinology, S025, Stanford University School of Medicine, Stanford, California 94305-5103. E-mail: or
| | - Dachuan Dong
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Kathrin Hasbargen
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Stefanie Bittner
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Yuan Cortez
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Andrew S Greenberg
- Obesity and Metabolism Laboratory, Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Salman Azhar
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Fredric B Kraemer
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Palo Alto, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Correspondence: Fredric B. Kraemer, MD, or Wen-Jun Shen, PhD, Division of Endocrinology, S025, Stanford University School of Medicine, Stanford, California 94305-5103. E-mail: or
| |
Collapse
|
23
|
Skowron KJ, Booker K, Cheng C, Creed S, David BP, Lazzara PR, Lian A, Siddiqui Z, Speltz TE, Moore TW. Steroid receptor/coactivator binding inhibitors: An update. Mol Cell Endocrinol 2019; 493:110471. [PMID: 31163202 PMCID: PMC6645384 DOI: 10.1016/j.mce.2019.110471] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 12/14/2022]
Abstract
The purpose of this review is to highlight recent developments in small molecules and peptides that block the binding of coactivators to steroid receptors. These coactivator binding inhibitors bind at the coregulator binding groove, also known as Activation Function-2, rather than at the ligand-binding site of steroid receptors. Steroid receptors that have been targeted with coactivator binding inhibitors include the androgen receptor, estrogen receptor and progesterone receptor. Coactivator binding inhibitors may be useful in some cases of resistance to currently prescribed therapeutics. The scope of the review includes small-molecule and peptide coactivator binding inhibitors for steroid receptors, with a particular focus on recent compounds that have been assayed in cell-based models.
Collapse
Affiliation(s)
- Kornelia J Skowron
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA
| | - Kenneth Booker
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA
| | - Changfeng Cheng
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA
| | - Simone Creed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA
| | - Brian P David
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA
| | - Phillip R Lazzara
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA
| | - Amy Lian
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA
| | - Zamia Siddiqui
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA
| | - Thomas E Speltz
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA; Department of Chemistry, University of Chicago, 929 E. 57th Street, E547, Chicago, IL, 60637, USA
| | - Terry W Moore
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL, 60612, USA; University of Illinois Cancer Center, University of Illinois at Chicago, 1801 W. Taylor Street, Chicago, IL, 60612, USA.
| |
Collapse
|
24
|
Cheung YM, Ramchand SK, Yeo B, Grossmann M. Cardiometabolic Effects of Endocrine Treatment of Estrogen Receptor-Positive Early Breast Cancer. J Endocr Soc 2019; 3:1283-1301. [PMID: 31259291 PMCID: PMC6595530 DOI: 10.1210/js.2019-00096] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022] Open
Abstract
Estrogen receptor-positive early breast cancer is common and has a relatively good prognosis. It shares risk factors with cardiovascular disease, and cardiovascular disease is an important competing cause of mortality. Adjuvant endocrine therapy with aromatase inhibitors (requiring concomitant ovarian suppression in premenopausal women) or selective estrogen receptor modulators (usually tamoxifen) exert oncologic benefits by respectively inhibiting estradiol synthesis or breast estrogen receptor signaling. Aromatase inhibitors cause systemic estradiol depletion. Tamoxifen has mixed agonistic/antagonistic effects in a tissue-dependent fashion. Given that estrogens modulate cardiometabolic risk, a review of the effects of endocrine therapy on cardiometabolic outcomes is pertinent. The current, but limited, evidence suggests that tamoxifen treatment, although associated with increases in body fat, hepatic steatosis, serum triglycerides, and diabetes risk, modestly reduces low-density lipoprotein cholesterol and lipoprotein(a) and may have favorable effects on markers of subclinical atherosclerosis. Tamoxifen is associated with either no effect on, or a reduction in, cardiovascular events, and it is associated with an increase in venous thromboembolic events. Aromatase inhibitors, although fewer studies are available and often confounded by comparison with tamoxifen, have not been consistently associated with adverse changes in cardiometabolic risk factors or increases in cardiovascular events. Further clinical trials designed to evaluate cardiometabolic outcomes are needed to more accurately determine the effects of endocrine therapy on cardiovascular risks, to inform individualized decisions regarding choice and duration of endocrine therapy, and to implement evidence-based strategies to mitigate cardiometabolic risks. In the meantime, although breast cancer-specific evidence for benefit of lifestyle measures is available and recommended routinely, proactive monitoring and treatment of cardiovascular risk factors should follow general population recommendations.
Collapse
Affiliation(s)
- Yee-Ming Cheung
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria Australia
| | - Sabashini K Ramchand
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria Australia
| | - Belinda Yeo
- Department of Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
| | - Mathis Grossmann
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria Australia
| |
Collapse
|
25
|
Chang YH, Ding DC, Chu TY. Estradiol and Progesterone Induced Differentiation and Increased Stemness Gene Expression of Human Fallopian Tube Epithelial Cells. J Cancer 2019; 10:3028-3036. [PMID: 31281480 PMCID: PMC6590043 DOI: 10.7150/jca.30588] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 05/03/2019] [Indexed: 12/12/2022] Open
Abstract
Fallopian tube epithelial cells (FTECs) are thought to be the origin of epithelial ovarian cancer. However, the effect of the hormones on FTECs is unknown, and therefore, this study explored this effect. We successfully derived FTECs from the fallopian tube epithelial layer and treated them with estradiol and progesterone. Reverse transcription polymerase chain reaction was used to evaluate the gene expression of the FTECs' hormone receptors. Confocal and electron microscopy were used to evaluate the morphology of the FTECs after they were treated with hormones. Finally, quantitative PCR was used to evaluate the gene expression of the hormone-treated FTECs. The results showed that the FTECs exhibited cuboidal cell morphology and could be maintained at a constant proliferation rate. Furthermore, flow cytometry revealed that the FTECs expressed stem cell markers, such as SSEA3, SSEA4, and Lgr5. Moreover, the FTECs could express both estrogen and progesterone receptors. In a culture treated with 400 nM estrogen, the FTECs differentiated toward ciliated cells, whereas in a culture treated with estradiol or progesterone, the FTECs increased their expression of certain stem cell markers (SSEA3, SSEA4, and Aldh1) and stemness genes [Wnt (AXIN2, LGR5, LGR6, and OLFM4) and Notch (Hes1) signaling]. In conclusion, hormones may alter the gene expressions of FTECs, and these cells may provide new insights into how FTECs regenerate in response to hormones.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- Stem Cell Laboratory, Department of Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation; Hualien, Taiwan.,Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation; Tzu Chi University, Hualien, Taiwan
| | - Dah-Ching Ding
- Stem Cell Laboratory, Department of Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation; Hualien, Taiwan.,Department of Obstetrics and Gynecology, Hualien Tzu-Chi Hospital, Buddhist Tzu Chi Medical Foundation; Tzu Chi University, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University; Hualien, Taiwan
| | - Tang-Yuan Chu
- Department of Obstetrics and Gynecology, Hualien Tzu-Chi Hospital, Buddhist Tzu Chi Medical Foundation; Tzu Chi University, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University; Hualien, Taiwan
| |
Collapse
|
26
|
Liu Y, Wang X, Wang G, Yang Y, Yuan Y, Ouyang L. The past, present and future of potential small-molecule drugs targeting p53-MDM2/MDMX for cancer therapy. Eur J Med Chem 2019; 176:92-104. [PMID: 31100649 DOI: 10.1016/j.ejmech.2019.05.018] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/02/2019] [Accepted: 05/06/2019] [Indexed: 02/05/2023]
Abstract
The p53 gene, a well-known tumor suppressor gene, plays a crucial role in cell cycle regulation, DNA repair, cell differentiation, and apoptosis. MDM2 exerts p53-dependent activity mainly by binding to p53 protein to form MDM2-p53 negative feedback loop. In addition, MDM2 is involved in a number of pathways that regulate cell proliferation and apoptosis, playing a p53-independent role. The p53 binding domain of MDMX bind to p53 transcriptional activation domain, inhibiting the transcriptional activity of p53 on its downstream genes, but does not mediate the degradation of p53. The anti-tumor effect is exerted by inhibiting the interaction between the MDM2/MDMX protein and the p53 protein by a small-molecule or by restoring the activity of the p53 protein. This review describes in the structural features, biological functions and mechanisms of p53-MDM2/MDMX, and summarizes small-molecule targeting p53-MDM2/MDMX.
Collapse
Affiliation(s)
- Yao Liu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, West China Hospital, Sichuan University, China
| | - Xiaohui Wang
- Department of Pharmacy, Naval Authorities Clinic, Beijing, 100841, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, West China Hospital, Sichuan University, China
| | - Yushang Yang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, West China Hospital, Sichuan University, China
| | - Yong Yuan
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, West China Hospital, Sichuan University, China.
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, West China Hospital, Sichuan University, China.
| |
Collapse
|
27
|
Gajadeera N, Hanson RN. Review of fluorescent steroidal ligands for the estrogen receptor 1995-2018. Steroids 2019; 144:30-46. [PMID: 30738074 DOI: 10.1016/j.steroids.2019.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/10/2019] [Accepted: 02/04/2019] [Indexed: 12/17/2022]
Abstract
The development of fluorescent ligands for the estrogen receptor (ER) continues to be of interest. Over the past 20 years, most efforts have focused on appending an expanding variety of fluorophores to the B-, C- and D-rings of the steroidal scaffold. This review highlights the synthesis and evaluation of derivatives substituted primarily at the 6-, 7α- and 17α-positions, culminating with our recent work on 11β-substituted estradiols, and proposes an approach to new fluorescent imaging agents that retain high ER affinity.
Collapse
Affiliation(s)
- Nisal Gajadeera
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston MA02115-5000, United States
| | - Robert N Hanson
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston MA02115-5000, United States.
| |
Collapse
|
28
|
Hernández-Hernández KL, Tapia-Orozco N, Gimeno M, Espinosa-García AM, García-García JA, Araiza-Olivera D, Sánchez-Bartez F, Gracia-Mora I, Gutierrez-Aguilar M, García-Arrazola R. Exposure to bisphenol A: current levels from food intake are toxic to human cells. Mol Biol Rep 2019; 46:2555-2559. [PMID: 30734171 DOI: 10.1007/s11033-019-04666-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/30/2019] [Indexed: 01/12/2023]
Abstract
In the present work, cell lines of different origin were exposed to BPA levels from food intake reported elsewhere. Specifically, we used an in vitro assay to determine cytotoxicity of BPA in three cell lines: MCF7 (breast cancer), PC3 (prostate cancer) and 3T3-L1 (mouse fibroblast). Cytotoxic effects were observed at concentrations higher than 50 μg/mL which is above the involuntary exposure level of BPA described before in fresh, canned and frozen foods and beverages. Furthermore, medial inhibitory concentrations (IC50) of 85.17 μg/mL and 88.48 μg/mL were observed for PC3 and 3T3-L1, respectively, and a slightly lower IC50 of 64.67 μg/mL for MCF7. These results highlight BPA's toxicity potential at current levels from food intake. The cell line-dependent divergent response to BPA reported herein is discussed.
Collapse
Affiliation(s)
- Karla L Hernández-Hernández
- Departamento de Alimentos y Biotecnologia, Facultad de Quimica, Universidad Nacional Autónoma de Mexico, Ciudad Universitaria, CP 04510, Coyoacan, Mexico
| | - Natalia Tapia-Orozco
- Departamento de Alimentos y Biotecnologia, Facultad de Quimica, Universidad Nacional Autónoma de Mexico, Ciudad Universitaria, CP 04510, Coyoacan, Mexico
| | - Miquel Gimeno
- Departamento de Alimentos y Biotecnologia, Facultad de Quimica, Universidad Nacional Autónoma de Mexico, Ciudad Universitaria, CP 04510, Coyoacan, Mexico
| | | | | | - Daniela Araiza-Olivera
- Departamento de Quimica de Biomacromoleculas, Instituto de Quimica, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Francisco Sánchez-Bartez
- Departamento de Química Inorgánica y Nuclear, Facultad de Química, UNAM, Ciudad Universitaria, C.U, Coyoacán, Ciudad de Mexico, Mexico
| | - Isabel Gracia-Mora
- Departamento de Química Inorgánica y Nuclear, Facultad de Química, UNAM, Ciudad Universitaria, C.U, Coyoacán, Ciudad de Mexico, Mexico
| | - Manuel Gutierrez-Aguilar
- Departamento de Bioquimica, Facultad de Quimica, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Roeb García-Arrazola
- Departamento de Alimentos y Biotecnologia, Facultad de Quimica, Universidad Nacional Autónoma de Mexico, Ciudad Universitaria, CP 04510, Coyoacan, Mexico.
| |
Collapse
|
29
|
Nagaraju B, Kovvuri J, Kumar CG, Routhu SR, Shareef MA, Kadagathur M, Adiyala PR, Alavala S, Nagesh N, Kamal A. Synthesis and biological evaluation of pyrazole linked benzothiazole-β-naphthol derivatives as topoisomerase I inhibitors with DNA binding ability. Bioorg Med Chem 2019; 27:708-720. [PMID: 30679134 DOI: 10.1016/j.bmc.2019.01.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 11/17/2022]
Abstract
A series of new pyrazole linked benzothiazole-β-naphthol derivatives were designed and synthesized using a simple, efficient and ecofriendly route under catalyst-free conditions in good to excellent yields. These derivatives were evaluated for their cytotoxicity on selected human cancer cell lines. Among those, the derivatives 4j, 4k and 4l exhibited considerable cytotoxicity with IC50 values ranging between 4.63 and 5.54 µM against human cervical cancer cells (HeLa). Structure activity relationship was elucidated by varying different substituents on benzothiazoles and pyrazoles. Further, flow cytometric analysis revealed that these derivatives induced cell cycle arrest in G2/M phase and spectroscopic studies such as UV-visible, fluorescence and circular dichroism studies showed that these derivatives exhibited good DNA binding affinity. Additionally, these derivatives can effectively inhibit the topoisomerase I activity. Viscosity studies and molecular docking studies demonstrated that the derivatives bind with the minor groove of the DNA.
Collapse
Affiliation(s)
- Burri Nagaraju
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
| | - Jeshma Kovvuri
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
| | - C Ganesh Kumar
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India.
| | - Sunitha Rani Routhu
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India
| | - Md Adil Shareef
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
| | - Manasa Kadagathur
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India
| | - Praveen Reddy Adiyala
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India
| | - Sateesh Alavala
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India
| | - Narayana Nagesh
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India.
| | - Ahmed Kamal
- Medicinal Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India; School of Pharmaceutical Education and Research, (SPER) Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
30
|
Musavi SAA, Yamashita S, Fujihara T, Masaka H, Islam MR, Kim S, Gotoh T, Kawahara M, Tashiro K, Yamauchi N. Analysis of differentially expressed genes and the promoters in bovine endometrium throughout estrus cycle and early pregnancy. Anim Sci J 2018; 89:1609-1621. [PMID: 30182475 DOI: 10.1111/asj.13091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/17/2018] [Indexed: 01/09/2023]
Abstract
Endometrial gene expression is primarily regulated by the ovarian steroids and pregnancy recognition factors. This study was aimed to characterize differential expression genes (DEGs) in bovine endometrium together with the analysis of their promoter region. Bovine uteri at follicular stage (FS), luteal stage (LS), and implantation stage (IS) at Day 18 of pregnancy were collected. Total RNA extracted and prepared cDNA were then subjected to high-throughput sequencing. For promoter analysis, 1 kb upstream promoter region of each DEG was analyzed. The numbers of highly expressed DEGs were 496 and 597 at FS and LS, respectively. When compared the gene expression of IS with LS, 383 and 346 DEGs showed higher and lower expression at IS, respectively. It was also observed that 20-30 transcription factors (TFs) were included in each DEGs. In addition, promoter analyses estimated 150-160 TFs for each stage. DLX4 and interferon regulatory factor 4 (IRF4) at FS, and IRF5, IRF9, STAT1, and STAT2 at IS were in common to DEGs and estimated TFs, respectively. This study highlighted potential molecular mechanisms controlling endometrial function during estrus cycle and IS, which will further guide to better understand the endometrial functions in future studies.
Collapse
Affiliation(s)
- Sayed A A Musavi
- Department of Animal and Marine Bio Resource Sciences, Graduate School Kyushu University, Fukuoka, Japan.,Department of Animal Husbandry, Agriculture Faculty, Balkh University, Mazar-e-Sharif, Afghanistan
| | - Seiya Yamashita
- Department of Animal and Marine Bio Resource Sciences, Graduate School Kyushu University, Fukuoka, Japan
| | - Taisuke Fujihara
- Department of Animal and Marine Bio Resource Sciences, Graduate School Kyushu University, Fukuoka, Japan
| | - Hironori Masaka
- Department of Animal and Marine Bio Resource Sciences, Graduate School Kyushu University, Fukuoka, Japan
| | - Md Rashedul Islam
- Department of Genetics and Animal Breeding, Faculty of Veterinary and Animal Science, Hajee Mohammad Danesh Science and Technology University, Dinajpur, Bangladesh
| | - Sangwan Kim
- Laboratory of Molecular Gene Technics, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takafumi Gotoh
- Laboratory of Meat Science, Graduate School of Agriculture, Kagoshima University, Kagoshima, Japan
| | - Manabu Kawahara
- Department of Animal Science, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Kosuke Tashiro
- Laboratory of Molecular Gene Technics, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Nobuhiko Yamauchi
- Department of Animal and Marine Bio Resource Sciences, Graduate School Kyushu University, Fukuoka, Japan
| |
Collapse
|
31
|
Goel P, Alam O, Naim MJ, Nawaz F, Iqbal M, Alam MI. Recent advancement of piperidine moiety in treatment of cancer- A review. Eur J Med Chem 2018; 157:480-502. [DOI: 10.1016/j.ejmech.2018.08.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/26/2018] [Accepted: 08/04/2018] [Indexed: 12/23/2022]
|
32
|
Denley MCS, Gatford NJF, Sellers KJ, Srivastava DP. Estradiol and the Development of the Cerebral Cortex: An Unexpected Role? Front Neurosci 2018; 12:245. [PMID: 29887794 PMCID: PMC5981095 DOI: 10.3389/fnins.2018.00245] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/28/2018] [Indexed: 12/16/2022] Open
Abstract
The cerebral cortex undergoes rapid folding in an "inside-outside" manner during embryonic development resulting in the establishment of six discrete cortical layers. This unique cytoarchitecture occurs via the coordinated processes of neurogenesis and cell migration. In addition, these processes are fine-tuned by a number of extracellular cues, which exert their effects by regulating intracellular signaling pathways. Interestingly, multiple brain regions have been shown to develop in a sexually dimorphic manner. In many cases, estrogens have been demonstrated to play an integral role in mediating these sexual dimorphisms in both males and females. Indeed, 17β-estradiol, the main biologically active estrogen, plays a critical organizational role during early brain development and has been shown to be pivotal in the sexually dimorphic development and regulation of the neural circuitry underlying sex-typical and socio-aggressive behaviors in males and females. However, whether and how estrogens, and 17β-estradiol in particular, regulate the development of the cerebral cortex is less well understood. In this review, we outline the evidence that estrogens are not only present but are engaged and regulate molecular machinery required for the fine-tuning of processes central to the cortex. We discuss how estrogens are thought to regulate the function of key molecular players and signaling pathways involved in corticogenesis, and where possible, highlight if these processes are sexually dimorphic. Collectively, we hope this review highlights the need to consider how estrogens may influence the development of brain regions directly involved in the sex-typical and socio-aggressive behaviors as well as development of sexually dimorphic regions such as the cerebral cortex.
Collapse
Affiliation(s)
- Matthew C. S. Denley
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Nicholas J. F. Gatford
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Katherine J. Sellers
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| |
Collapse
|
33
|
Zhang HB, Wang XD, Xu K, Li XG. The progress of prophylactic treatment in retinopathy of prematurity. Int J Ophthalmol 2018; 11:858-873. [PMID: 29862189 DOI: 10.18240/ijo.2018.05.24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/28/2017] [Indexed: 12/19/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a retinal vascular disorder frequently found in premature infants. Different therapeutic strategies have been developed to treat ROP. However, there are still many children with ROP suffering by severe limitations in vision or even blindness. Recently, ROP has been suggested to be caused by abnormal development of the retinal vasculature, but not simply resulted by retinal neovascularization which takes about 4 to 6wk after birth in premature infants. Thus, instead of focusing on how to reduce retinal neovascularization, understanding the pathological changes and mechanisms that occur prior to retinal neovascularization is meaningful, which may lead to identify novel target(s) for the development of novel strategy to promote the healthy growth of retinal blood vessels rather than passively waiting for the appearance of retinal neovascularization and removing it by force. In this review, we discussed recent studies about, 1) the pathogenesis prior to retinal neovascularization in oxygen-induced retinopathy (OIR; a ROP in animal model) and in premature infants with ROP; 2) the preclinical and clinical research on preventive treatment of early OIR and ROP. We will not only highlight the importance of the mechanisms and signalling pathways in regulating early stage of ROP but also will provide guidance for actively exploring novel mechanisms and discovering novel treatments for early phase OIR and ROP prior to retinal neovascularization in the future.
Collapse
Affiliation(s)
- Hong-Bing Zhang
- Eye Institute of Shaanxi Province; Xi'an First Hospital, Xi'an 710002, Shaanxi Province, China
| | - Xiao-Dong Wang
- Eye Institute of Shaanxi Province; Xi'an First Hospital, Xi'an 710002, Shaanxi Province, China
| | - Kun Xu
- Eye Institute of Shaanxi Province; Xi'an First Hospital, Xi'an 710002, Shaanxi Province, China
| | - Xiao-Gang Li
- Department of Internal Medicine; Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
34
|
Kharman-Biz A, Gao H, Ghiasvand R, Haldosen LA, Zendehdel K. Expression of the three components of linear ubiquitin assembly complex in breast cancer. PLoS One 2018; 13:e0197183. [PMID: 29763465 PMCID: PMC5953448 DOI: 10.1371/journal.pone.0197183] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 04/27/2018] [Indexed: 11/19/2022] Open
Abstract
Proteins belonging to the linear ubiquitin assembly complex (LUBAC) are believed to be important in tumorigenesis. LUBAC has been demonstrated to be composed of RBCK1, RNF31 and SHARPIN. The aim of this study was to explore all members of the LUBAC complex as novel biomarkers in breast cancer. We have already reported that RNF31 mRNA levels are higher in breast cancer samples compared to adjacent non-tumor tissue. In this study we extend these findings by demonstrating that the mRNA levels of RBCK1 and SHARPIN are also higher in tumors compared to adjacent non-tumor tissue in the same cross sectional study of samples (p < 0.001). In addition, up-regulated mRNA expression of all three members of the LUBAC complex displayed high predictive value in distinguishing tumor tissues from adjacent non-tumor tissue as determined by ROC curve analysis. Furthermore, we investigated whether there is an association between the mRNA and protein expression levels of RBCK1, RNF31 and SHARPIN and clinicopathological parameters including estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor (HER2) status and found that RNF31 protein is significantly higher in ERalpha-negative tumors than ERalpha-positive tumors (p = 0.034). Collectively, our findings indicate that up-regulated mRNA expression of RNF31, RBCK1 and SHARPIN could potentially be diagnostic biomarkers of breast cancer and RNF31 might be a drug target for ERalpha-negative breast cancers.
Collapse
Affiliation(s)
- Amirhossein Kharman-Biz
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Reza Ghiasvand
- Oslo Centre for Biostatistics and Epidemiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Lars-Arne Haldosen
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Kazem Zendehdel
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
- * E-mail:
| |
Collapse
|
35
|
Stefkovich ML, Arao Y, Hamilton KJ, Korach KS. Experimental models for evaluating non-genomic estrogen signaling. Steroids 2018; 133:34-37. [PMID: 29122548 PMCID: PMC5864539 DOI: 10.1016/j.steroids.2017.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/02/2017] [Indexed: 10/18/2022]
Abstract
Non-genomic effects of estrogen receptor α (ERα) signaling have been described for decades. However, the mechanisms and physiological processes resulting solely from non-genomic signaling are poorly understood. Challenges in studying these effects arise from the strongly nucleophilic tendencies of estrogen receptor, and many approaches to excluding ERα from the nucleus have been explored over the years. In this review, we discuss past strategies for studying ERα's non-genomic action and current models, specifically H2NES ERα, first described by Burns et al. (2011). In vitro and preliminary in vivo data from H2NES ERα and H2NES mice suggest a promising avenue for pinpointing specific non-genomic ERα action.
Collapse
Affiliation(s)
- Megan L Stefkovich
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institutes of Health, NIEHS, 111 TW Alexander Dr, Research Triangle Park, NC 27709, USA
| | - Yukitomo Arao
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institutes of Health, NIEHS, 111 TW Alexander Dr, Research Triangle Park, NC 27709, USA
| | - Katherine J Hamilton
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institutes of Health, NIEHS, 111 TW Alexander Dr, Research Triangle Park, NC 27709, USA
| | - Kenneth S Korach
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institutes of Health, NIEHS, 111 TW Alexander Dr, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
36
|
Catanese MC, Vandenberg LN. Developmental estrogen exposures and disruptions to maternal behavior and brain: Effects of ethinyl estradiol, a common positive control. Horm Behav 2018; 101:113-124. [PMID: 29107581 PMCID: PMC5938171 DOI: 10.1016/j.yhbeh.2017.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/28/2017] [Accepted: 10/24/2017] [Indexed: 12/22/2022]
Abstract
Due of its structural similarity to the endogenous estrogen 17β-estradiol (E2), the synthetic estrogen 17α-ethinyl estradiol (EE2) is widely used to study the effects of estrogenic substances on sensitive organs at multiple stages of development. Here, we investigated the effects of EE2 on maternal behavior and the maternal brain in females exposed during gestation and the perinatal period. We assessed several components of maternal behavior including nesting behavior and pup retrieval; characterized the expression of estrogen receptor (ER)α in the medial preoptic area (MPOA), a brain region critical for the display of maternal behavior; and measured expression of tyrosine hydroxylase, a marker for dopaminergic cells, in the ventral tegmental area (VTA), a brain region important in maternal motivation. We found that developmental exposure to EE2 induces subtle effects on several aspects of maternal behavior including time building the nest and time spent engaged in self-care. Developmental exposure to EE2 also altered ERα expression in the central MPOA during both early and late lactation and led to significantly reduced tyrosine hydroxylase immunoreactivity in the VTA. Our results demonstrate both dose- and postpartum stage-related effects of developmental exposure to EE2 on behavior and brain that manifest later in adulthood, during the maternal period. These findings provide further evidence for effects of exposure to exogenous estrogenic compounds during the critical periods of fetal and perinatal development.
Collapse
Affiliation(s)
- Mary C Catanese
- Program in Neuroscience and Behavior, University of Massachusetts - Amherst, USA
| | - Laura N Vandenberg
- Program in Neuroscience and Behavior, University of Massachusetts - Amherst, USA; Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts - Amherst, USA.
| |
Collapse
|
37
|
Marchal E, Figliola C, Thompson A. Prodigiosenes conjugated to tamoxifen and estradiol. Org Biomol Chem 2018. [PMID: 28628182 DOI: 10.1039/c7ob00943g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We report the synthesis of the first click-appended prodigiosene conjugates. Four prodigiosene conjugates of estradiol functionalised at the 7α-position were prepared, as were three prodigiosene conjugates of tamoxifen. The coupling between a prodigiosene and an 11-hydroxy estradiol derivative via an ether linkage was investigated, as was the 11- and 7-functionalisation of the estradiol core. The robustness of estradiol protecting groups was severely challenged by reactions typically used to equip such frameworks for 11- and 7-functionalisation. Specifically, and important to synthesis involving estradiol, TBS, TMS and THP are not useful protecting groups for the functionalisation of this core. When the chemical features of the therapeutic agent limit the choice of protecting group (in this case, prodigiosenes bearing aryl, NH, alkenyl and ester groups), click chemistry becomes an attractive synthetic strategy. The anti-cancer activity of the seven click prodigiosene conjugates was evaluated.
Collapse
Affiliation(s)
- Estelle Marchal
- Department of Chemistry, Dalhousie University, PO BOX 15000, Halifax, NS B3H 4R2, Canada.
| | | | | |
Collapse
|
38
|
The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget 2018; 8:8921-8946. [PMID: 27888811 PMCID: PMC5352454 DOI: 10.18632/oncotarget.13475] [Citation(s) in RCA: 378] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 10/13/2016] [Indexed: 01/10/2023] Open
Abstract
Cancer has long been a grievous disease complicated by innumerable players aggravating its cure. Many clinical studies demonstrated the prognostic relevance of the tumor suppressor protein p53 for many human tumor types. Overexpression of mutated p53 with reduced or abolished function is often connected to resistance to standard medications, including cisplatin, alkylating agents (temozolomide), anthracyclines, (doxorubicin), antimetabolites (gemcitabine), antiestrogenes (tamoxifen) and EGFR-inhibitors (cetuximab). Such mutations in the TP53 gene are often accompanied by changes in the conformation of the p53 protein. Small molecules that restore the wild-type conformation of p53 and, consequently, rebuild its proper function have been identified. These promising agents include PRIMA-1, MIRA-1, and several derivatives of the thiosemicarbazone family. In addition to mutations in p53 itself, p53 activity may be also be impaired due to alterations in p53s regulating proteins such as MDM2. MDM2 functions as primary cellular p53 inhibitor and deregulation of the MDM2/p53-balance has serious consequences. MDM2 alterations often result in its overexpression and therefore promote inhibition of p53 activity. To deal with this problem, a judicious approach is to employ MDM2 inhibitors. Several promising MDM2 inhibitors have been described such as nutlins, benzodiazepinediones or spiro-oxindoles as well as novel compound classes such as xanthone derivatives and trisubstituted aminothiophenes. Furthermore, even naturally derived inhibitor compounds such as a-mangostin, gambogic acid and siladenoserinols have been discovered. In this review, we discuss in detail such small molecules that play a pertinent role in affecting the p53-MDM2 signaling axis and analyze their potential as cancer chemotherapeutics.
Collapse
|
39
|
Al-Khyatt W, Tufarelli C, Khan R, Iftikhar SY. Selective oestrogen receptor antagonists inhibit oesophageal cancer cell proliferation in vitro. BMC Cancer 2018; 18:121. [PMID: 29390981 PMCID: PMC5796348 DOI: 10.1186/s12885-018-4030-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 01/23/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Oestrogen receptors (ER) have a well-established role to the initiation, progression and regulation of responses to treatment of breast, prostate, and lung cancers. Previous data indicates altered ER expression in oesophageal cancers (OC). However the role of ER subtypes and ER specific inhibitors in the regulation of OC progression remains unclear. This study sought to assess levels of ERα and ERβ in OC. The effects of highly selective ER antagonists on cell proliferation and apoptosis in two OC adenocarcinoma cell lines was also studied. METHODS ERα and ERβ expression profiling in paired normal oesophageal mucosa and tumour tissues (n = 34; adenocarcinoma n = 28; squamous cell carcinoma n = 6) was performed using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Correlation between levels of ER with the clinico-pathological features for OC was determined. The effect of selective ER antagonists on proliferation of OE33 and OE19 OC cell lines was studied. RESULTS ERα and ERβ mRNA expression was significantly higher (p < 0.05) in tumour tissues relative to their paired normal mucosa and correlated inversely with survival outcome (p < 0.05). Upregulation of ERα mRNA correlated with higher pathological T-stage (p < 0.05) and lymph node metastasis (p < 0.05) while ERβ mRNA upregulation correlated with positive vascular invasion (p < 0.05). A significant concentration-dependent inhibition of proliferation in OE33 and OE19 cell lines was induced by a highly-selective ERα antagonist (MPP) and an ERβ specific antagonist (PHTPP) (p < 0.05). Moreover, anti-oestrogens induced cell death through stimulation of apoptotic caspase activity. CONCLUSION These findings indicate that the ER system is involved in OC progression and thus may provide a novel target for the treatment of OC.
Collapse
Affiliation(s)
- Waleed Al-Khyatt
- Department of Upper GI Surgery, Royal Derby Hospital, Derby Teaching Hospitals NHS Foundation Trust, Uttoxeter Road, Derby, DE22 3NE UK
- Division of Medical Sciences and Graduate Entry Medicine, Royal Derby Hospital, Uttoxeter Road, Derby, DE22 3DT UK
| | - Cristina Tufarelli
- Division of Medical Sciences and Graduate Entry Medicine, Royal Derby Hospital, Uttoxeter Road, Derby, DE22 3DT UK
| | - Raheela Khan
- Division of Medical Sciences and Graduate Entry Medicine, Royal Derby Hospital, Uttoxeter Road, Derby, DE22 3DT UK
| | - Syed Yousef Iftikhar
- Department of Upper GI Surgery, Royal Derby Hospital, Derby Teaching Hospitals NHS Foundation Trust, Uttoxeter Road, Derby, DE22 3NE UK
- Division of Medical Sciences and Graduate Entry Medicine, Royal Derby Hospital, Uttoxeter Road, Derby, DE22 3DT UK
| |
Collapse
|
40
|
Sukocheva OA. Expansion of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Function in Normal and Cancer Cells: From Membrane Restructuring to Mediation of Estrogen Signaling and Stem Cell Programming. Int J Mol Sci 2018; 19:ijms19020420. [PMID: 29385066 PMCID: PMC5855642 DOI: 10.3390/ijms19020420] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 02/05/2023] Open
Abstract
Sphingolipids, sphingolipid metabolizing enzymes, and their receptors network are being recognized as part of the signaling mechanisms, which govern breast cancer cell growth, migration, and survival during chemotherapy treatment. Approximately 70% of breast cancers are estrogen receptor (ER) positive and, thus, rely on estrogen signaling. Estrogen activates an intracellular network composed of many cytoplasmic and nuclear mediators. Some estrogen effects can be mediated by sphingolipids. Estrogen activates sphingosine kinase 1 (SphK1) and amplifies the intracellular concentration of sphingosine-1-phosphate (S1P) in breast cancer cells during stimulation of proliferation and survival. Specifically, Estrogen activates S1P receptors (S1PR) and induces growth factor receptor transactivation. SphK, S1P, and S1PR expression are causally associated with endocrine resistance and progression to advanced tumor stages in ER-positive breast cancers in vivo. Recently, the network of SphK/S1PR was shown to promote the development of ER-negative cancers and breast cancer stem cells, as well as stimulating angiogenesis. Novel findings confirm and broaden our knowledge about the cross-talk between sphingolipids and estrogen network in normal and malignant cells. Current S1PRs therapeutic inhibition was indicated as a promising chemotherapy approach in non-responsive and advanced malignancies. Considering that sphingolipid signaling has a prominent role in terminally differentiated cells, the impact should be considered when designing specific SphK/S1PR inhibitors. This study analyzes the dynamic of the transformation of sphingolipid axis during a transition from normal to pathological condition on the level of the whole organism. The sphingolipid-based mediation and facilitation of global effects of estrogen were critically accented as a bridging mechanism that should be explored in cancer prevention.
Collapse
Affiliation(s)
- Olga A Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park, SA 5042, Australia.
| |
Collapse
|
41
|
Perkins MS, Louw-du Toit R, Africander D. A comparative characterization of estrogens used in hormone therapy via estrogen receptor (ER)-α and -β. J Steroid Biochem Mol Biol 2017; 174:27-39. [PMID: 28743541 DOI: 10.1016/j.jsbmb.2017.07.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/10/2017] [Accepted: 07/18/2017] [Indexed: 02/07/2023]
Abstract
Conventional hormone therapy (HT) containing estrogens such as ethinylestradiol (EE) have been associated with an increased risk of breast cancer and cardiovascular disease resulting in women seeking safer alternatives that are claimed to have fewer health risks. One such alternative gaining popularity, is custom-compounded bioidentical (b)HT formulations containing bioidentical estradiol (bE2) and estriol (bE3). However, the preparation of these custom-compounded estrogens is not regulated, and depending on the route of synthesis, steroid mixtures with differing activities may be produced. Thus, an investigation into the activities of estrogens prepared by custom-compounded pharmacies is warranted. The aim of this study was therefore to directly compare the pharmacological properties of bE2 and bE3 of unknown purity relative to commercially available, pure E2, E3 and estrone (E1) standards as well as synthetic EE used in conventional HT via the human estrogen receptor (ER)-α and -β. We determined precise equilibrium dissociation constants (Kd or Ki values) and showed that bE2 and bE3 display similar binding affinities to the E2 and E3 standards, while EE had a higher affinity for ERα, and E1 a lower affinity for ERβ. Furthermore, all the estrogens display similar agonist efficacies, but not potencies, for transactivation on a minimal ERE-containing promoter via the individual ER subtypes. Although E2 and E3 were equally efficacious and potent on the endogenous ERE-containing pS2 promoter in the MCF-7 BUS breast cancer cell line co-expressing ERα and ERβ, E1 was less efficacious and potent than E2. This study is the first to demonstrate that the bioidentical estrogens, commercially available estrogen standards and synthetic EE are full agonists for transrepression on both minimal and endogenous NFκB-containing promoters. Moreover, we showed that these estrogens all increase proliferation and anchorage-independent growth of MCF-7 BUS cells to a similar extent, suggesting that custom-compounded bHT may in fact not be a safer alternative to conventional HT. Furthermore, our results showing that E3 and E1 are not weak estrogens, and that E3 does not antagonize the activity of E2, suggest that the rationale behind the use of E3 and E1 in custom-compounded bHT formulations should be readdressed. Taken together, the results indicating that there is mostly no difference between the custom-compounded bioidentical estrogens, commercially available estrogen standards and synthetic EE, at concentrations reflecting serum levels in women using estrogen-containing HT, suggest that there is no clear advantage in choosing bHT above conventional HT.
Collapse
Affiliation(s)
- Meghan S Perkins
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland 7602, South Africa.
| | - Renate Louw-du Toit
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland 7602, South Africa.
| | - Donita Africander
- Department of Biochemistry, Stellenbosch University, Private Bag X1, Matieland 7602, South Africa.
| |
Collapse
|
42
|
Hu Z, Shen WJ, Kraemer FB, Azhar S. Regulation of adrenal and ovarian steroidogenesis by miR-132. J Mol Endocrinol 2017; 59:269-283. [PMID: 28729436 PMCID: PMC6376965 DOI: 10.1530/jme-17-0011] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022]
Abstract
miR-132 is hormonally regulated in steroidogenic cells of the adrenal gland, ovary and testis. Here, we examined the potential role of miR-132 in the control of steroidogenesis. Transfection of Y1 adrenal cells with miR-132 increased mRNAs of 3β-HSD and 20α-HSD enzymes, which catalyze the sequential conversion of pregnenolone to progesterone to biologically inactive 20α-hydroxyprogesterone (20α-OHP). Overexpression of miR-132 reduced MeCP2 and StAR protein expression, basal progestin (progesterone and 20α-OHP) production, but enhanced their production in response to cAMP stimulation. Use of [3H] pregnenolone and free-diffusible 22(R)-hydroxycholesterol further confirmed that miR-132 promotes the production of 20α-OHP by upregulating 3β-HSD and 20α-HSD. Evidence is also presented that StAR is a direct target of miR-132. Transient transfection of Y1 cells with miR-132 demonstrated that miR-132 induction of 3β-HSD and 20α-HSD was accompanied by significant suppression of one of its target gene products, MeCP2. In contrast, co-expression of miR-132 plus MeCP2 protein partially blocked the ability of miR-132 to upregulate the expression and function of 3β-HSD and 20α-HSD. Moreover, suppression of MeCP2 protein with siRNA resulted in increased expression of 3β-HSD and 20α-HSD, further demonstrating that miR-132 induces the expression of these two enzymes via inhibition of MeCP2. Likewise, overexpression of miR-132 increased 20α-OHP production with and without HDL loading, while knockdown of miR-132 resulted in a significant decrease of 20α-OHP production by granulosa cells. In conclusion, our data suggest that miR-132 attenuates steroidogenesis by repressing StAR expression and inducing 20α-HSD via inhibition of MeCP2 to generate a biologically inactive 20α-OHP.
Collapse
Affiliation(s)
- Zhigang Hu
- Geriatric ResearchEducation and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Division of EndocrinologyGerontology and Metabolism, Stanford University, Stanford, California, USA
| | - Wen-Jun Shen
- Geriatric ResearchEducation and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Division of EndocrinologyGerontology and Metabolism, Stanford University, Stanford, California, USA
| | - Fredric B Kraemer
- Geriatric ResearchEducation and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Division of EndocrinologyGerontology and Metabolism, Stanford University, Stanford, California, USA
| | - Salman Azhar
- Geriatric ResearchEducation and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Division of EndocrinologyGerontology and Metabolism, Stanford University, Stanford, California, USA
| |
Collapse
|
43
|
Boivin FJ, Schmidt-Ott KM. Transcriptional mechanisms coordinating tight junction assembly during epithelial differentiation. Ann N Y Acad Sci 2017. [PMID: 28636799 DOI: 10.1111/nyas.13367] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epithelial tissues form a selective barrier via direct cell-cell interactions to separate and establish concentration gradients between the different compartments of the body. Proper function and formation of this barrier rely on the establishment of distinct intercellular junction complexes. These complexes include tight junctions, adherens junctions, desmosomes, and gap junctions. The tight junction is by far the most diverse junctional complex in the epithelial barrier. Its composition varies greatly across different epithelial tissues to confer various barrier properties. Thus, epithelial cells rely on tightly regulated transcriptional mechanisms to ensure proper formation of the epithelial barrier and to achieve tight junction diversity. Here, we review different transcriptional mechanisms utilized during embryogenesis and disease development to promote tight junction assembly and maintenance of intercellular barrier integrity. We focus particularly on the Grainyhead-like transcription factors and ligand-activated nuclear hormone receptors, two central families of proteins in epithelialization.
Collapse
Affiliation(s)
- Felix J Boivin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology, Charité Medical University, Berlin, Germany
| |
Collapse
|
44
|
Miller MM, McMullen PD, Andersen ME, Clewell RA. Multiple receptors shape the estrogen response pathway and are critical considerations for the future of in vitro-based risk assessment efforts. Crit Rev Toxicol 2017; 47:564-580. [DOI: 10.1080/10408444.2017.1289150] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
45
|
Zhang S, Wang Z, Hu Z, Li C, Tang C, Carlson KE, Luo J, Dong C, Katzenellenbogen JA, Huang J, Zhou HB. Selenophenes: Introducing a New Element into the Core of Non-Steroidal Estrogen Receptor Ligands. ChemMedChem 2017; 12:235-249. [PMID: 27976818 DOI: 10.1002/cmdc.201600593] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Indexed: 02/06/2023]
Abstract
The importance of the heterocyclic core elements with peripheral phenolic and alkyl substituents as a dominant structural motif of ligands for the estrogen receptor (ER) has been well recognized. In this study we expanded the structural diversity of core elements by preparing selenium-containing heterocycles and exploring the activities of these selenophenes on the two ERs, ERα and ERβ. Careful structure-activity relationship (SAR) analysis of their ER binding affinities showed that most selenophenes are ERβ-selective, with the position of the phenol substituents on the selenophene core and the nature of these substituents having a marked effect on their binding affinities. The compound bis(2-fluoro-4-hydroxyphenyl)selenophene (2 f) has the highest relative binding affinity (RBA) of 24.3 for ERβ. In transcription assays, most selenophenes were found to exhibit partial to full agonist activity for both ER subtypes, with compounds bis(2-methyl-4-hydroxyphenyl)selenophene (2 b), bis(4-fluoro-3-hydroxyphenyl)3-bromoselenophene (6 f), and 2,3,5-tris(hydroxyphenyl)thiophenes (8 b and 8 d) profiling as superagonists for ERα; however, several compounds display a range of ERα or ERβ antagonistic activities. A few selenophenes exhibited antiproliferative activity, with compound 8 c showing antiproliferative effects similar to that of 4-hydroxytamoxifen in breast cancer MCF-7 cells while being nontoxic to normal VERO cells. These new ligands could act as models for the development of novel agents leading to improved therapeutics that target the estrogen receptor.
Collapse
Affiliation(s)
- Silong Zhang
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 185 East Lake Road, Wuhan, 430071, P.R. China
| | - Zhiyong Wang
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 185 East Lake Road, Wuhan, 430071, P.R. China
| | - Zhiye Hu
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 185 East Lake Road, Wuhan, 430071, P.R. China
| | - Changhao Li
- College of Life Sciences, Wuhan University, No. 299 Bayi Road, Wuhan, 430072, P.R. China
| | - Chu Tang
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 185 East Lake Road, Wuhan, 430071, P.R. China
| | - Kathryn E Carlson
- Department of Chemistry, University of Illinois, 600 South Mathews Avenue, Urbana, IL, 61801, USA
| | - Junjie Luo
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 185 East Lake Road, Wuhan, 430071, P.R. China
| | - Chune Dong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 185 East Lake Road, Wuhan, 430071, P.R. China
| | - John A Katzenellenbogen
- Department of Chemistry, University of Illinois, 600 South Mathews Avenue, Urbana, IL, 61801, USA
| | - Jian Huang
- College of Life Sciences, Wuhan University, No. 299 Bayi Road, Wuhan, 430072, P.R. China
| | - Hai-Bing Zhou
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 185 East Lake Road, Wuhan, 430071, P.R. China.,Institute of Pharmacy & Pharmacology, University of South China, Hengyang, 421001, China
| |
Collapse
|
46
|
Vaquero J, Nguyen Ho-Bouldoires TH, Clapéron A, Fouassier L. Role of the PDZ-scaffold protein NHERF1/EBP50 in cancer biology: from signaling regulation to clinical relevance. Oncogene 2017; 36:3067-3079. [PMID: 28068322 DOI: 10.1038/onc.2016.462] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/26/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022]
Abstract
The transmission of cellular information requires fine and subtle regulation of proteins that need to interact in a coordinated and specific way to form efficient signaling networks. The spatial and temporal coordination relies on scaffold proteins. Thanks to protein interaction domains such as PDZ domains, scaffold proteins organize multiprotein complexes enabling the proper transmission of cellular information through intracellular networks. NHERF1/EBP50 is a PDZ-scaffold protein that was initially identified as an organizer and regulator of transporters and channels at the apical side of epithelia through actin-binding ezrin-moesin-radixin proteins. Since, NHERF1/EBP50 has emerged as a major regulator of cancer signaling network by assembling cancer-related proteins. The PDZ-scaffold EBP50 carries either anti-tumor or pro-tumor functions, two antinomic functions dictated by EBP50 expression or subcellular localization. The dual function of NHERF1/EBP50 encompasses the regulation of several major signaling pathways engaged in cancer, including the receptor tyrosine kinases PDGFR and EGFR, PI3K/PTEN/AKT and Wnt-β-catenin pathways.
Collapse
Affiliation(s)
- J Vaquero
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - T H Nguyen Ho-Bouldoires
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - A Clapéron
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - L Fouassier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|
47
|
Zaitseva M, Yue DS, Katzenellenbogen JA, Rogers PAW, Gargett CE. Estrogen Receptor-α Agonists Promote Angiogenesis in Human Myometrial Microvascular Endothelial Cells. ACTA ACUST UNITED AC 2016; 11:529-35. [PMID: 15582497 DOI: 10.1016/j.jsgi.2004.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The relative role of the two estrogen receptors, ERalpha and ERbeta, in mediating angiogenic responses in adult human endothelium is unknown. The aim of this study was to determine whether novel ERalpha-selective agonists, propyl pyrazole triol (PPT) and the tetrahydrochrysene (R,R-THC), up-regulate the expression of vascular endothelial growth factor receptor-2 (VEGFR-2), and promote VEGF-stimulated endothelial cell proliferation in primary cultures of adult female microvascular endothelial cells co-expressing endogenous ERalpha and ERbeta. METHODS Confluent primary cultures of microvascular endothelial cells isolated from human myometrium were incubated with 17beta-estradiol (1 and 10 nM), PPT (10 nM to 3 microM), or R,R-THC (10 nM to 3 microM) for 18 hours and VEGFR-2 expression measured by biotin-VEGF165 binding and flow cytometry. Endothelial cell proliferation was assessed in microvascular endothelial cells after incubation with 17beta-estradiol (10 nM), PPT (100 nM), and R,R-THC (100 nM) for 6 days using a tetrazolium-based bioassay. RESULTS Both PPT and R,R-THC increased VEGFR-2 expression on myometrial microvascular endothelial cells in a dose-dependent manner, reaching a maximum at 1 microM. Approximately 40% of myometrial microvascular endothelial cell isolates only express ERbeta and do not express ERalpha, and in these neither PPT, R,R-THC, nor 17beta-estradiol increased VEGF binding. PPT- or R,R-THC-stimulated increase in VEGF binding was significantly different between ERalpha+ and ERalpha- microvascular endothelial cell samples (P < .001 and P < .05, respectively). PPT, R,R-THC, and 17beta-estradiol significantly augmented VEGF-stimulated microvascular endothelial cell proliferation in ERalpha+ (P < .05), but not in ERalpha- samples. CONCLUSIONS This angiogenic effect of 17beta-estradiol on adult female microvascular endothelial cells is mediated by ERalpha, rather than ERbeta.
Collapse
Affiliation(s)
- Marina Zaitseva
- Centre for Women's Health Research, Monash University Department of Obstetrics and Gynaecology, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | | | | | |
Collapse
|
48
|
Niu AQ, Xie LJ, Wang H, Zhu B, Wang SQ. Prediction of selective estrogen receptor beta agonist using open data and machine learning approach. Drug Des Devel Ther 2016; 10:2323-31. [PMID: 27486309 PMCID: PMC4958355 DOI: 10.2147/dddt.s110603] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Estrogen receptors (ERs) are nuclear transcription factors that are involved in the regulation of many complex physiological processes in humans. ERs have been validated as important drug targets for the treatment of various diseases, including breast cancer, ovarian cancer, osteoporosis, and cardiovascular disease. ERs have two subtypes, ER-α and ER-β. Emerging data suggest that the development of subtype-selective ligands that specifically target ER-β could be a more optimal approach to elicit beneficial estrogen-like activities and reduce side effects. Methods Herein, we focused on ER-β and developed its in silico quantitative structure-activity relationship models using machine learning (ML) methods. Results The chemical structures and ER-β bioactivity data were extracted from public chemogenomics databases. Four types of popular fingerprint generation methods including MACCS fingerprint, PubChem fingerprint, 2D atom pairs, and Chemistry Development Kit extended fingerprint were used as descriptors. Four ML methods including Naïve Bayesian classifier, k-nearest neighbor, random forest, and support vector machine were used to train the models. The range of classification accuracies was 77.10% to 88.34%, and the range of area under the ROC (receiver operating characteristic) curve values was 0.8151 to 0.9475, evaluated by the 5-fold cross-validation. Comparison analysis suggests that both the random forest and the support vector machine are superior for the classification of selective ER-β agonists. Chemistry Development Kit extended fingerprints and MACCS fingerprint performed better in structural representation between active and inactive agonists. Conclusion These results demonstrate that combining the fingerprint and ML approaches leads to robust ER-β agonist prediction models, which are potentially applicable to the identification of selective ER-β agonists.
Collapse
Affiliation(s)
- Ai-Qin Niu
- Department of Gynecology, the First People's Hospital of Shangqiu, Shangqiu, Henan, People's Republic of China
| | - Liang-Jun Xie
- Department of Image Diagnoses, the Third Hospital of Jinan, Jinan, Shandong, People's Republic of China
| | - Hui Wang
- Department of Gynecology, the First People's Hospital of Shangqiu, Shangqiu, Henan, People's Republic of China
| | - Bing Zhu
- Department of Gynecology, the First People's Hospital of Shangqiu, Shangqiu, Henan, People's Republic of China
| | - Sheng-Qi Wang
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| |
Collapse
|
49
|
Cocci P, Palermo FA, Quassinti L, Bramucci M, Miano A, Mosconi G. Determination of estrogenic activity in the river Chienti (Marche Region, Italy) by using in vivo and in vitro bioassays. J Environ Sci (China) 2016; 43:48-53. [PMID: 27155408 DOI: 10.1016/j.jes.2015.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/18/2015] [Accepted: 07/18/2015] [Indexed: 06/05/2023]
Abstract
Environmental estrogen-like compounds (i.e. xenoestrogens) are a variety of pollutants, ranging from synthetic to natural occurring molecules, that are found in surface and waste waters over a wide range of concentrations. In aquatic environment, the overall estrogenic activity is often due to the presence of a mixture of chemicals and their degraded products which can induce synergistic effects. Current strategies for monitoring estrogen-like chemicals are based on the use of a battery of in vivo and in vitro ecotoxicological tests. In this regard, the aim of the present work was to carry out a bio-monitoring study for testing estrogenicity of the Chienti river (Marche Region, Italy) by using both an E-screen and a vitellogenin (Vtg) induction assay in juvenile goldfish. Three sites were used for analysis, localized at the mouth (sampling point 1), in the middle (sampling point 2) and at the origin (sampling point 3) of Chienti river. For most of the water samples (i.e. samples collected at sampling points 2 and 3), clear estrogenic activity was detected in the E-screen assay suggesting different proliferation activities in function of the collecting site. In contrast, the Vtg ELISA demonstrated that water samples collected from each sampling point were estrogenic. Overall, we showed for the first time that the estrogenic activities in water samples from the Chienti river were significant in both in vivo and in vitro; we also observed a different sensitivity between bioassays.
Collapse
Affiliation(s)
- Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy
| | - Francesco Alessandro Palermo
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy.
| | - Luana Quassinti
- School of Pharmacy, University of Camerino, I-62032 Camerino, MC, Italy
| | - Massimo Bramucci
- School of Pharmacy, University of Camerino, I-62032 Camerino, MC, Italy
| | - Antonino Miano
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy
| | - Gilberto Mosconi
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy
| |
Collapse
|
50
|
Sato M, Ohta K, Kaise A, Aoto S, Endo Y. Symmetric 4,4'-(piperidin-4-ylidenemethylene)bisphenol derivatives as novel tunable estrogen receptor (ER) modulators. Bioorg Med Chem 2016; 24:1089-94. [PMID: 26822566 DOI: 10.1016/j.bmc.2016.01.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 01/14/2016] [Accepted: 01/18/2016] [Indexed: 11/15/2022]
Abstract
We designed and synthesized 4,4'-(piperidin-4-ylidenemethylene)bisphenol derivatives as novel tunable estrogen receptor (ER) modulators. The introduction of hydrophobic substituents on the nitrogen atom of the piperidine ring enhanced ERα binding affinity. In addition, the introduction of four methyl groups adjacent to the piperidine ring nitrogen atom remarkably enhanced ERα binding affinity. N-Acetyl-2,2,6,6-tetramethylpiperidine derivative 3b showed high ERα binding affinity, high MCF-7 cell proliferation inducing activity, and high metabolic stability in rat liver S9 fractions.
Collapse
Affiliation(s)
- Manabu Sato
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Kiminori Ohta
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan.
| | - Asako Kaise
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Sayaka Aoto
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Yasuyuki Endo
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| |
Collapse
|