1
|
Yu R. Exaggerated Increases in the Serum Cortisol Level in a Woman Following Oral Contraceptive Treatment. AACE Clin Case Rep 2024; 10:206-209. [PMID: 39372822 PMCID: PMC11447753 DOI: 10.1016/j.aace.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 10/08/2024] Open
Abstract
Background/Objective Extreme hypercortisolemia in an otherwise healthy patient can be due to familial dysalbuminemia, generalized glucocorticoid resistance, and estrogen-containing medications. I report a woman who appeared to have an exaggerated increase in the serum cortisol level following oral contraceptive treatment. Case Report A 50-year-old woman presented with extreme morning hypercortisolemia-cortisol levels of 61 and 55 mcg/dL (4 and 3 months before presentation, respectively; normal range, 8-25 mcg/dL)-found during workup of mildly increased white cell counts. The morning cortisol level had been 10 mcg/dL after administration of 1-mg dexamethasone. The 24-hour urine free cortisol level had been normal and only slightly increased after correction by creatinine. The patient was anxious about the extremely high cortisol levels but otherwise felt well. She took norgestimate-ethinyl estradiol contraceptive (0.18/0.215/0.25 mg - 35 mcg). Physical examination showed a well-appearing, lean female. The thyroid-stimulating hormone, total thyroxine, free thyroxine, total triiodothyronine, free triiodothyronine, androstenedione, dehydroepiandrosterone sulfate, aldosterone, and renin levels were normal. Morning total cortisol and cortisol-binding globulin (CBG) were tested before and after she held the oral contraceptive for 2 months. The total cortisol and CBG levels decreased from 50 to 26 mcg/dL and from 6.4 to 3.8 mg/dL (normal range, 1.7-3.1 mg/dL), respectively. Discussion Increases in the serum cortisol-binding proteins are a well-recognized cause for increases in the serum cortisol levels. Conclusion This case suggests that modern oral contraceptives with low to moderate estrogen activity can cause extreme increases in the serum cortisol levels due to marked increases in the CBG levels.
Collapse
Affiliation(s)
- Run Yu
- Division of Endocrinology, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, California
| |
Collapse
|
2
|
Sluka P, Ackermann U, Rigopoulos A, Wardan H, Pezaro C, Burvenich IJ, Scott AM, Davis ID. Characterization of an Estrogen Receptor α-Selective 18 F-Estradiol PET Tracer. World J Nucl Med 2024; 23:153-160. [PMID: 39170834 PMCID: PMC11335392 DOI: 10.1055/s-0044-1786518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
Objective Conventional imaging of cancer with modalities such as computed tomography or magnetic resonance imaging provides little information about the underlying biology of the cancer and consequently little guidance for systemic treatment choices. Accurate identification of aggressive cancers or those that are likely to respond to specific treatment regimens would allow more precisely tailored treatments to be used. The expression of the estrogen receptor α subunit is associated with a more aggressive phenotype, with a greater propensity to metastasize. We aimed to characterize the binding properties of an 18 F-estradiol positron emission tomography (PET) tracer in its ability to bind to the α and β forms of estrogen receptors in vitro and confirmed its binding to estrogen receptor α in vivo. Methods The 18 F-estradiol PET tracer was synthesized and its quality confirmed by high-performance liquid chromatography. Binding of the tracer was assessed in vitro by saturation and competitive binding studies to HEK293T cells transfected with estrogen receptor α ( ESR1 ) and/or estrogen receptor β ( ESR2 ). Binding of the tracer to estrogen receptor α in vivo was assessed by imaging of uptake of the tracer into MCF7 xenografts in BALB/c nu/nu mice. Results The 18 F-estradiol PET tracer bound with high affinity (94 nM) to estrogen receptor α, with negligible binding to estrogen receptor β. Uptake of the tracer was observed in MCF7 xenografts, which almost exclusively express estrogen receptor α. Conclusion 18 F-estradiol PET tracer binds in vitro with high specificity to the estrogen receptor α isoform, with minimal binding to estrogen receptor β. This may help distinguish human cancers with biological dependence on estrogen receptor subtypes.
Collapse
Affiliation(s)
- Pavel Sluka
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
| | - Uwe Ackermann
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
- The University of Melbourne, Parkville, VIC, Australia
| | - Angela Rigopoulos
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Hady Wardan
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
| | - Carmel Pezaro
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- Department of Oncology, Eastern Health, Box Hill, VIC, Australia
| | - Ingrid J.G. Burvenich
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Andrew M. Scott
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
- The University of Melbourne, Parkville, VIC, Australia
| | - Ian D. Davis
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- Department of Oncology, Eastern Health, Box Hill, VIC, Australia
| |
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
|
Cimmino A, Fasciglione GF, Gioia M, Marini S, Ciaccio C. Multi-Anticancer Activities of Phytoestrogens in Human Osteosarcoma. Int J Mol Sci 2023; 24:13344. [PMID: 37686148 PMCID: PMC10487502 DOI: 10.3390/ijms241713344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Phytoestrogens are plant-derived bioactive compounds with estrogen-like properties. Their potential health benefits, especially in cancer prevention and treatment, have been a subject of considerable research in the past decade. Phytoestrogens exert their effects, at least in part, through interactions with estrogen receptors (ERs), mimicking or inhibiting the actions of natural estrogens. Recently, there has been growing interest in exploring the impact of phytoestrogens on osteosarcoma (OS), a type of bone malignancy that primarily affects children and young adults and is currently presenting limited treatment options. Considering the critical role of the estrogen/ERs axis in bone development and growth, the modulation of ERs has emerged as a highly promising approach in the treatment of OS. This review provides an extensive overview of current literature on the effects of phytoestrogens on human OS models. It delves into the multiple mechanisms through which these molecules regulate the cell cycle, apoptosis, and key pathways implicated in the growth and progression of OS, including ER signaling. Moreover, potential interactions between phytoestrogens and conventional chemotherapy agents commonly used in OS treatment will be examined. Understanding the impact of these compounds in OS holds great promise for developing novel therapeutic approaches that can augment current OS treatment modalities.
Collapse
Affiliation(s)
| | | | | | | | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, I-00133 Rome, Italy; (A.C.); (G.F.F.); (M.G.); (S.M.)
| |
Collapse
|
5
|
Impacts of synthetic androgen and estrogenic antagonist administration on growth performance, sex steroids hormones, and immune markers of male and female broilers. Poult Sci 2022; 102:102244. [PMID: 36335738 PMCID: PMC9640317 DOI: 10.1016/j.psj.2022.102244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/23/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The influence of synthetic androgen and estrogenic antagonists (Tamoxifen) on body characteristics and immune response of male and female broilers and the correlation between sex hormone levels were estimated in our experiment. One day old chicks were sexed, and chicks of each sex were randomly distributed on three experimental treatments; the first treatment group (TAM20) chicks were supplied with estrogenic antagonist tamoxifen citrate 20 mg/kg body weight through oral administration for four times every other day from third until ninth d; Androgen treatment chicks were injected intramuscular with veterinary androgen AD GAN@ (Boldenone Undecylenate 50 mg) 1 cm/10 kg body weight at fifth and ninth day, and the third treatment was control. Androgen treatment reported the highest feed intake with the lowest for TAM20 treatment. Concerning carcass characteristics, early androgen injection increased breast percentage significantly compared to TAM20 treatment. Androgen supplementation increased significantly comb the percentage. However, TAM20 decreased it particularly compared to control. Moreover, the percentage of comb and shanks was substantially higher for males than females. Concerning the effects of both treatments on sex hormones, androgen showed favorable effects on testosterone and estrogen compared to Tamoxifen 20 treatment. On the other hand, the administration of TAM 20 improves phagocytic activity compared to androgen administration.
Collapse
|
6
|
FitzPatrick AM. Is Estrogen a Missing Culprit in Thyroid Eye Disease? Sex Steroid Hormone Homeostasis Is Key to Other Fibrogenic Autoimmune Diseases - Why Not This One? Front Immunol 2022; 13:898138. [PMID: 35784325 PMCID: PMC9248759 DOI: 10.3389/fimmu.2022.898138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Sex bias in autoimmune disease (AID) prevalence is known, but the role of estrogen in disease progression is more complex. Estrogen can even be protective in some AIDs; but in systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and systemic sclerosis (SSc), estrogen, its metabolites, and its receptors have been demonstrated to play critical, localized inflammatory roles. Estrogen is instrumental to the fibrosis seen in RA, SLE, SSc and other disease states, including breast cancer and uterine leiomyomas. Fibrotic diseases tend to share a common pattern in which lymphocyte-monocyte interactions generate cytokines which stimulate the deposition of fibrogenic connective tissue. RA, SLE, SSc and thyroid eye disease (TED) have very similar inflammatory and fibrotic patterns-from pathways to tissue type. The thorough investigations that demonstrated estrogen's role in the pathology of RA, SLE, and SSc could, and possibly should, be carried out in TED. One might even expect to find an even greater role for estrogen, and sex steroid homeostasis in TED, given that TED is typically sequalae to Graves' disease (GD), or Hashimoto's disease (HD), and these are endocrine disorders that can create considerable sex steroid hormone dysregulation. This paper highlights the pathophysiology similarities in 4 AIDs, examines the evidence of sex steroid mediated pathology across 3 AIDs and offers a case study and speculation on how this may be germane to TED.
Collapse
|
7
|
Wu G, Zhou J, Zhu X, Tang X, Liu J, Zhou Q, Chen Z, Liu T, Wang W, Xiao X, Wu T. Integrative analysis of expression, prognostic significance and immune infiltration of RFC family genes in human sarcoma. Aging (Albany NY) 2022; 14:3705-3719. [PMID: 35483337 PMCID: PMC9085243 DOI: 10.18632/aging.204039] [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: 02/08/2022] [Accepted: 04/13/2022] [Indexed: 11/25/2022]
Abstract
Objective: To reveal the expression and prognostic value of replication factor C family genes (RFCs) in patients with sarcoma. Results: The results showed that the mRNA expression levels of RFC2, RFC3, RFC4, and RFC5 were increased in sarcoma tissues. In addition, Cancer Cell Line Encyclopedia (CCLE) dataset analysis indicated that RFC1, RFC2, RFC3, RFC4, and RFC5 were elevated expressed in sarcoma cell lines. Moreover, Gene Expression Profiling Interactive Analysis (GEPIA) and Kaplan-Meier Plotter showed that highly expressed RFC2-5 were associated with poor overall survival (OS) or relapse-free survival (RFS) in sarcoma patients. The results of the Tumor Immune Estimation Resource (TIMER) database indicated that the expression of RFCs was negatively correlated with the infiltration of CD4+ T cells and macrophages. Conclusions: There were significant differences in the expression of RFCs between normal tissue and sarcoma tissue, and RFC2, RFC3, RFC4, and RFC5 might be promising prognostic biomarkers for sarcoma. Methods: The expression of RFCs was analyzed using the ONCOMINE dataset and GEPIA dataset. CCLE dataset was used to assess the expression of RFCs in the cancer cell line. The prognostic value of RFCs was evaluated by GEPIA and Kaplan-Meier analysis. Furthermore, the association between RFCs and their co-expressed genes were explored via ONCOMINE and GEPIA datasets. We used the TIMER dataset to analyze the immune cell infiltration of RFCs in sarcoma.
Collapse
Affiliation(s)
- Gen Wu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.,Clinical Medicine Eight-Year Program, 02 Class, 2014 Grade, Central South University, Changsha 410013, Hunan Province, China
| | - Jian Zhou
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xi Zhu
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich 81377, Germany
| | - Xianzhe Tang
- Department of Orthopedics, Chenzhou No.1 People's Hospital, Chenzhou 423000, Hunan, China
| | - Jie Liu
- Department of Cardiology, The Fourth Hospital of Changsha, Changsha 410006, Hunan, China
| | - Qiong Zhou
- Department of Cardiology, The Fourth Hospital of Changsha, Changsha 410006, Hunan, China
| | - Ziyuan Chen
- Department of Orthopedics, The First People's Hospital of Changde City, Changde 415003, Hunan, China
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Wanchun Wang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xungang Xiao
- Department of Orthopedics, Chenzhou No.1 People's Hospital, Chenzhou 423000, Hunan, China
| | - Tong Wu
- Department of Emergency, The First Hospital of Changsha, Changsha 410005, Hunan, China
| |
Collapse
|
8
|
Trott JF, Schennink A, Horigan KC, Lemay DG, Cohen JR, Famula TR, Dragon JA, Hovey RC. Unique Transcriptomic Changes Underlie Hormonal Interactions During Mammary Histomorphogenesis in Female Pigs. Endocrinology 2022; 163:bqab256. [PMID: 34918063 PMCID: PMC10409904 DOI: 10.1210/endocr/bqab256] [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: 09/28/2021] [Indexed: 11/19/2022]
Abstract
Successful lactation and the risk for developing breast cancer depend on growth and differentiation of the mammary gland (MG) epithelium that is regulated by ovarian steroids (17β-estradiol [E] and progesterone [P]) and pituitary-derived prolactin (PRL). Given that the MG of pigs share histomorphogenic features present in the normal human breast, we sought to define the transcriptional responses within the MG of pigs following exposure to all combinations of these hormones. Hormone-ablated female pigs were administered combinations of E, medroxyprogesterone 17-acetate (source of P), and either haloperidol (to induce PRL) or 2-bromo-α-ergocryptine. We subsequently monitored phenotypic changes in the MG including mitosis, receptors for E and P (ESR1 and PGR), level of phosphorylated STAT5 (pSTAT5), and the frequency of terminal ductal lobular unit (TDLU) subtypes; these changes were then associated with all transcriptomic changes. Estrogen altered the expression of approximately 20% of all genes that were mostly associated with mitosis, whereas PRL stimulated elements of fatty acid metabolism and an inflammatory response. Several outcomes, including increased pSTAT5, highlighted the ability of E to enhance PRL action. Regression of transcriptomic changes against several MG phenotypes revealed 1669 genes correlated with proliferation, among which 29 were E inducible. Additional gene expression signatures were associated with TDLU formation and the frequency of ESR1 or PGR. These data provide a link between the hormone-regulated genome and phenome of the MG in a species having a complex histoarchitecture like that in the human breast, and highlight an underexplored synergy between the actions of E and PRL during MG development.
Collapse
Affiliation(s)
- Josephine F Trott
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| | - Anke Schennink
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| | - Katherine C Horigan
- Department of Animal Science, University of Vermont, Burlington, Vermont 05405, USA
| | - Danielle G Lemay
- US Department of Agriculture ARS Western Human Nutrition Research Center, Davis, California 95616, USA
| | - Julia R Cohen
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| | - Thomas R Famula
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| | - Julie A Dragon
- Vermont Integrative Genomics Resource, University of Vermont, Burlington, Vermont 05405, USA
| | - Russell C Hovey
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| |
Collapse
|
9
|
Rurak GM, Simard S, Freitas-Andrade M, Lacoste B, Charih F, Van Geel A, Stead J, Woodside B, Green JR, Coppola G, Salmaso N. Sex differences in developmental patterns of neocortical astroglia: A mouse translatome database. Cell Rep 2022; 38:110310. [PMID: 35108542 DOI: 10.1016/j.celrep.2022.110310] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/22/2021] [Accepted: 01/06/2022] [Indexed: 02/08/2023] Open
Abstract
Astroglial cells are key players in the development and maintenance of neurons and neuronal networks. Astroglia express steroid hormone receptors and show rapid responses to hormonal manipulations. However, despite important sex differences in the cortex and hippocampus, few studies have examined sex differences in astroglial cells in telencephalic development. To characterize the cortical astroglial translatome in male and female mice across postnatal development, we use translating ribosome affinity purification together with RNA sequencing and immunohistochemistry to phenotype astroglia at six developmental time points. Overall, we find two distinct astroglial phenotypes between early (P1-P7) and late development (P14-adult), independent of sex. We also find sex differences in gene expression patterns across development that peak at P7 and appear to result from males reaching a mature astroglial phenotype earlier than females. These developmental sex differences could have an impact on the construction of neuronal networks and windows of vulnerability to perturbations and disease.
Collapse
Affiliation(s)
- Gareth M Rurak
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Stephanie Simard
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Moises Freitas-Andrade
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - François Charih
- Department of Systems and Computer Engineering, Carleton University, Ottawa, ON, Canada
| | - Amanda Van Geel
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - John Stead
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada; Concordia University, Montreal, QC, Canada
| | - James R Green
- Department of Systems and Computer Engineering, Carleton University, Ottawa, ON, Canada
| | - Gianfilippo Coppola
- Department of Pathology, Yale University, 333 Cedar St., New Haven, CT 06511, USA.
| | - Natalina Salmaso
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada; Department of Pathology, Yale University, 333 Cedar St., New Haven, CT 06511, USA.
| |
Collapse
|
10
|
Mori H, Saeki K, Chang G, Wang J, Wu X, Hsu PY, Kanaya N, Wang X, Somlo G, Nakamura M, Bild A, Chen S. Influence of Estrogen Treatment on ESR1+ and ESR1- Cells in ER + Breast Cancer: Insights from Single-Cell Analysis of Patient-Derived Xenograft Models. Cancers (Basel) 2021; 13:cancers13246375. [PMID: 34944995 PMCID: PMC8699443 DOI: 10.3390/cancers13246375] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary The benefit of endocrine therapy is normally observed for cancers with 10% or more of cells positive for ER expression. We compared the gene expression profiles in both ESR1+ and ESR1– cells in ER+ tumors following estrogen treatment. Our single-cell RNA sequencing analysis of estrogen-stimulated (SC31) and estrogen-suppressed (GS3) patient-derived xenograft models offered an unprecedented opportunity to address the molecular and functional differences between ESR1+ and ESR1– cells. While estrogen should activate ERα and stimulate ESR1+ cells, our findings regarding ESR1– cells were important, indicating that the proliferation of ESR1– cells in ER+ cancer is also influenced by estrogen. Another valuable finding from our studies was that estrogen also upregulated a tumor-suppressor gene, IL-24, only in GS3. Estrogen increased the percentage of cells expressing IL-24, associated with the estrogen-dependent inhibition of GS3 tumor growth. Abstract A 100% ER positivity is not required for an endocrine therapy response. Furthermore, while estrogen typically promotes the progression of hormone-dependent breast cancer via the activation of estrogen receptor (ER)-α, estrogen-induced tumor suppression in ER+ breast cancer has been clinically observed. With the success in establishing estrogen-stimulated (SC31) and estrogen-suppressed (GS3) patient-derived xenograft (PDX) models, single-cell RNA sequencing analysis was performed to determine the impact of estrogen on ESR1+ and ESR1– tumor cells. We found that 17β-estradiol (E2)-induced suppression of GS3 transpired through wild-type and unamplified ERα. E2 upregulated the expression of estrogen-dependent genes in both SC31 and GS3; however, E2 induced cell cycle advance in SC31, while it resulted in cell cycle arrest in GS3. Importantly, these gene expression changes occurred in both ESR1+ and ESR1– cells within the same breast tumors, demonstrating for the first time a differential effect of estrogen on ESR1– cells. E2 also upregulated a tumor-suppressor gene, IL-24, in GS3. The apoptosis gene set was upregulated and the G2M checkpoint gene set was downregulated in most IL-24+ cells after E2 treatment. In summary, estrogen affected pathologically defined ER+ tumors differently, influencing both ESR1+ and ESR1– cells. Our results also suggest IL-24 to be a potential marker of estrogen-suppressed tumors.
Collapse
Affiliation(s)
- Hitomi Mori
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
- Department of Surgery and Oncology, Graduate School of Medicine, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Kohei Saeki
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - Gregory Chang
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - Jinhui Wang
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, 655 Huntington Drive, Monrovia, CA 91016, USA; (J.W.); (X.W.)
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, 655 Huntington Drive, Monrovia, CA 91016, USA; (J.W.); (X.W.)
| | - Pei-Yin Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - Xiaoqiang Wang
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - George Somlo
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA 91010, USA; (G.S.); (A.B.)
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medicine, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Andrea Bild
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA 91010, USA; (G.S.); (A.B.)
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
- Correspondence: ; Tel.: +1-626-218-3454; Fax: +1-626-301-8972
| |
Collapse
|
11
|
Khaksari M, Hajializadeh Z, Mahani SE, Soltani Z, Asadikaram G. Estrogen receptor agonists induce anti‑edema effects by altering α and β estrogen receptor gene expression. Acta Neurobiol Exp (Wars) 2021; 81:286-294. [PMID: 34672299 DOI: 10.21307/ane-2021-027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The present study aimed to examine whether the attenuation of estrogen receptor expression is prevented by propyl pyrazole triol (PPT), an agonist for estrogen receptor α (ERα) or and diarypropiolnitrile (DPN), an agonist for estrogen receptor β (ERβ) after traumatic brain injury (TBI). The tests performed on ovariectomized female Wistar rats included sham group, vehicle group, and treated groups: PPT, DPN, and PPT+DPN 30 minutes after TBI. Blood‑brain barrier (BBB) disruption and brain water content were estimated. RT‑PCR and\r\nwestern blotting were utilized to evaluate ESR1 and ESR2 gene and protein expression. The data indicated that PPT, DPN, and PPT+DPN attenuated TBI‑induced brain edema. Also, BBB disruption after TBI was prevented in PPT, DPN, and PPT+DPN‑treated TBI animals. Estrogen agonist‑treated animals showed a significant elevation in Esr1 mRNA and protein expression levels in the brain tissue of TBI rats. In addition, the data indicated a significant elevation of Esr2 mRNA and protein expression levels in the brain tissue of estrogen agonist‑treated TBI rats. The data shows that both ESR1 and ESR2 agonists can enhance ER mRNA and protein levels in TBI animals' brain. It appears that this effect contributes to the neuroprotective function of ER agonists.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran;
| | - Zahra Hajializadeh
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Esmaeili Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Asadikaram
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
12
|
Moga MA, Dimienescu OG, Bălan A, Dima L, Toma SI, Bîgiu NF, Blidaru A. Pharmacological and Therapeutic Properties of Punica granatum Phytochemicals: Possible Roles in Breast Cancer. Molecules 2021; 26:molecules26041054. [PMID: 33671442 PMCID: PMC7921999 DOI: 10.3390/molecules26041054] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Pomgranate (Punica granatum) represents a high source of polyphenols with great bioavailability. The role of this fruit in the prevention and treatment of various malignant pathologies has been long time cited in both scientific and non-scientific literature, making thus important to identify its involvement in the pathophysiological processes. The treatment for breast cancer had focused on the inhibition of the mechanisms that governs the estrogen activity. These mechanisms are covered either by the antagonism of the estrogen receptor (ER) or by the inhibition of the estrogen synthesis. Our interest in identifying a bioactive compound rich in polyphenols, which induces both the antagonism of the estrogen receptor, and the inhibition of the estrogen synthesis, revealed us the pomegranate fruit and its derivatives: peel and seeds. Pomegranates' chemical composition include many biological active substances such as flavonols, flavanols, anthocyanins, proanthocyanidins, ellagitannins and gallotannins. Materials and Methods: We performed a review of the scientific literature by using the following keywords: "pomegranate", "breast cancer", "Punica granatum", "pomegranate polyphenols". Our search was performed in the PubMed and Google Scholar databases, and it included only original research written in English from the last 20 years. None of the articles were excluded due to affiliation. A total number of 28 original papers, which mentioned the beneficial activity of pomegranate against breast cancer, were selected. Both clinical and preclinical studies were considered for this review. Results: Recent discoveries pointed out that polyphenols from Punica granatum possess strong anti-cancer activity, exhibited by a variety of mechanisms, such as anti-estrogenic, anti-proliferative, anti-angiogenetic, anti-inflammatory, and anti-metastatic. Pomegranate extracts induced cell cycle arrest in the G0/G1 phase, and induced cytotoxicity in a dose- and time-dependent manner. Moreover, several polyphenols extracted from pomegranate inhibited the invasion potential, migration and viability of breast cancer cells. The effects of pomegranate juice on serum estrogens and other sexual hormones levels were also investigated on two human cohorts. Conclusions: Punica granatum represents a promising area in oncology. The large availability and low cost, associated with the lack of side effects, made from this natural product a great strategy for the management of breast cancer. There are several mechanistic studies in mouse models and in breast cancer cell lines, suggesting the possible pathways through which polyphenols from pomegranate extracts act, but larger and better-controlled studies are necessary in the future. Only two small clinical trials were conducted on humans until now, but their results are contradictory and should be considered preliminary.
Collapse
Affiliation(s)
- Marius Alexandru Moga
- Department of Medical and Surgical Specialities, Faculty of Medicine, Transilvania University of Brasov, 500032 Brasov, Romania; (M.A.M.); (O.G.D.); (A.B.)
| | - Oana Gabriela Dimienescu
- Department of Medical and Surgical Specialities, Faculty of Medicine, Transilvania University of Brasov, 500032 Brasov, Romania; (M.A.M.); (O.G.D.); (A.B.)
| | - Andreea Bălan
- Department of Medical and Surgical Specialities, Faculty of Medicine, Transilvania University of Brasov, 500032 Brasov, Romania; (M.A.M.); (O.G.D.); (A.B.)
| | - Lorena Dima
- Department of Fundamental, Prophylactical and Clinical Disciplines, Faculty of Medicine, Transilvania University of Brasov, 500032 Brasov, Romania; (L.D.); (S.I.T.)
| | - Sebastian Ionut Toma
- Department of Fundamental, Prophylactical and Clinical Disciplines, Faculty of Medicine, Transilvania University of Brasov, 500032 Brasov, Romania; (L.D.); (S.I.T.)
| | - Nicușor Florin Bîgiu
- Department of Medical and Surgical Specialities, Faculty of Medicine, Transilvania University of Brasov, 500032 Brasov, Romania; (M.A.M.); (O.G.D.); (A.B.)
- Correspondence: ; Tel.: +40-728519031
| | - Alexandru Blidaru
- Department of Surgical Oncology, Oncological Institute “Al. Trestioneanu” of Bucharest, University of Medicine and Pharmacy Carol Davila, 020021 Bucharest, Romania;
| |
Collapse
|
13
|
Mal R, Magner A, David J, Datta J, Vallabhaneni M, Kassem M, Manouchehri J, Willingham N, Stover D, Vandeusen J, Sardesai S, Williams N, Wesolowski R, Lustberg M, Ganju RK, Ramaswamy B, Cherian MA. Estrogen Receptor Beta (ERβ): A Ligand Activated Tumor Suppressor. Front Oncol 2020; 10:587386. [PMID: 33194742 PMCID: PMC7645238 DOI: 10.3389/fonc.2020.587386] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) belong to a superfamily of nuclear receptors called steroid hormone receptors, which, upon binding ligand, dimerize and translocate to the nucleus where they activate or repress the transcription of a large number of genes, thus modulating critical physiologic processes. ERβ has multiple isoforms that show differing association with prognosis. Expression levels of the full length ERβ1 isoform are often lower in aggressive cancers as compared to normal tissue. High ERβ1 expression is associated with improved overall survival in women with breast cancer. The promise of ERβ activation, as a potential targeted therapy, is based on concurrent activation of multiple tumor suppressor pathways with few side effects compared to chemotherapy. Thus, ERβ is a nuclear receptor with broad-spectrum tumor suppressor activity, which could serve as a potential treatment target in a variety of human cancers including breast cancer. Further development of highly selective agonists that lack ERα agonist activity, will be necessary to fully harness the potential of ERβ.
Collapse
Affiliation(s)
- Rahul Mal
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Alexa Magner
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Joel David
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Jharna Datta
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Meghna Vallabhaneni
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Mahmoud Kassem
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Jasmine Manouchehri
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Natalie Willingham
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Daniel Stover
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Jeffery Vandeusen
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Sagar Sardesai
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Nicole Williams
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Robert Wesolowski
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Maryam Lustberg
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Ramesh K Ganju
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Bhuvaneswari Ramaswamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Mathew A Cherian
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
14
|
Ayyar VS, Jusko WJ. Transitioning from Basic toward Systems Pharmacodynamic Models: Lessons from Corticosteroids. Pharmacol Rev 2020; 72:414-438. [PMID: 32123034 DOI: 10.1124/pr.119.018101] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Technology in bioanalysis, -omics, and computation have evolved over the past half century to allow for comprehensive assessments of the molecular to whole body pharmacology of diverse corticosteroids. Such studies have advanced pharmacokinetic and pharmacodynamic (PK/PD) concepts and models that often generalize across various classes of drugs. These models encompass the "pillars" of pharmacology, namely PK and target drug exposure, the mass-law interactions of drugs with receptors/targets, and the consequent turnover and homeostatic control of genes, biomarkers, physiologic responses, and disease symptoms. Pharmacokinetic methodology utilizes noncompartmental, compartmental, reversible, physiologic [full physiologically based pharmacokinetic (PBPK) and minimal PBPK], and target-mediated drug disposition models using a growing array of pharmacometric considerations and software. Basic PK/PD models have emerged (simple direct, biophase, slow receptor binding, indirect response, irreversible, turnover with inactivation, and transduction models) that place emphasis on parsimony, are mechanistic in nature, and serve as highly useful "top-down" methods of quantitating the actions of diverse drugs. These are often components of more complex quantitative systems pharmacology (QSP) models that explain the array of responses to various drugs, including corticosteroids. Progressively deeper mechanistic appreciation of PBPK, drug-target interactions, and systems physiology from the molecular (genomic, proteomic, metabolomic) to cellular to whole body levels provides the foundation for enhanced PK/PD to comprehensive QSP models. Our research based on cell, animal, clinical, and theoretical studies with corticosteroids have provided ideas and quantitative methods that have broadly advanced the fields of PK/PD and QSP modeling and illustrates the transition toward a global, systems understanding of actions of diverse drugs. SIGNIFICANCE STATEMENT: Over the past half century, pharmacokinetics (PK) and pharmacokinetics/pharmacodynamics (PK/PD) have evolved to provide an array of mechanism-based models that help quantitate the disposition and actions of most drugs. We describe how many basic PK and PK/PD model components were identified and often applied to the diverse properties of corticosteroids (CS). The CS have complications in disposition and a wide array of simple receptor-to complex gene-mediated actions in multiple organs. Continued assessments of such complexities have offered opportunities to develop models ranging from simple PK to enhanced PK/PD to quantitative systems pharmacology (QSP) that help explain therapeutic and adverse CS effects. Concurrent development of state-of-the-art PK, PK/PD, and QSP models are described alongside experimental studies that revealed diverse CS actions.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Department of Pharmaceutical Sciences University at Buffalo, School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| | - William J Jusko
- Department of Pharmaceutical Sciences University at Buffalo, School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| |
Collapse
|
15
|
Ayyar VS, DuBois DC, Almon RR, Jusko WJ. Modeling Corticosteroid Pharmacokinetics and Pharmacodynamics, Part III: Estrous Cycle and Estrogen Receptor-Dependent Antagonism of Glucocorticoid-Induced Leucine Zipper (GILZ) Enhancement by Corticosteroids. J Pharmacol Exp Ther 2019; 370:337-349. [PMID: 31197018 DOI: 10.1124/jpet.119.257543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/10/2019] [Indexed: 01/04/2023] Open
Abstract
Our previous report examined the pharmacokinetics (PK) of methylprednisolone (MPL) and adrenal suppression after a 50 mg/kg IM bolus in male and female rats, and we described in detail the development of a minimal physiologically based pharmacokinetic/pharmacodynamic (mPBPK/PD) model. In continuation of such assessments, we investigated sex differences in genomic MPL responses (PD). Message expression of the glucocorticoid-induced leucine zipper (GILZ) was chosen as a multitissue biomarker of glucocorticoid receptor (GR)-mediated drug response. Potential time-dependent interplay between sex hormone and glucocorticoid signaling in vivo was assessed by comparing the enhancement of GILZ by MPL in the uterus [high estrogen receptor (ER) density] and in liver (lower ER density) from male and female rats dosed within the proestrus (high estradiol/progesterone) and estrus (low estradiol/progesterone) phases of the rodent estrous cycle. An expanded-systems PD model of MPL considering circadian rhythms, multireceptor (ER and GR) control, and estrous variations delineated the determinants controlling receptor/gene-mediated steroid responses. Hepatic GILZ response was ∼3-fold greater in females, regardless of estrous stage, compared with males, driven predominantly by increased MPL exposure in females and a negligible influence of estrogen interaction. In contrast, GILZ response in the uterus during proestrus in females was 60% of that observed in estrus-phased females, despite no PK or receptor differences, providing in vivo support to the hypothesis of estrogen-mediated antagonism of glucocorticoid signaling. The developed model offers a mechanistic platform to assess the determinants of sex and tissue specificity in corticosteroid actions and, in turn, reveals a unique PD drug-hormone interaction occurring in vivo. SIGNIFICANCE STATEMENT: Mechanisms relating to sex-based pharmacodynamic variability in genomic responses to corticosteroids have been unclear. Using combined experimental and systems pharmacology modeling approaches, sex differences in both pharmacokinetic and pharmacodynamic mechanisms controlling the enhancement of a sensitive corticosteroid-regulated biomarker, the glucocorticoid-induced leucine zipper (GILZ), were clarified in vivo. The multiscale minimal physiologically based pharmacokinetics/pharmacodynamic model successfully captured the experimental observations and quantitatively discerned the roles of the rodent estrous cycle (hormonal variation) and tissue specificity in mediating the antagonistic coregulation of GILZ gene synthesis. These findings collectively support the hypothesis that estrogens antagonize pharmacodynamic signaling of genomic corticosteroid actions in vivo in a time- and estrogen receptor-dependent manner.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York
| | - Debra C DuBois
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York
| | - Richard R Almon
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York
| | - William J Jusko
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
16
|
Roberts RD. Is Estrogen the Answer for Osteosarcoma? Cancer Res 2019; 79:1034-1035. [PMID: 30877098 DOI: 10.1158/0008-5472.can-19-0209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 11/16/2022]
Abstract
Bone biologists have long understood how estrogen drives osteoclasts to make new bone. While evidence hinted that a loss of estrogen signals might also play a role in bone-forming cancers like osteosarcoma, this idea has remained untested. Lillo Osuna and colleagues present data demonstrating widespread epigenetic silencing of estrogen receptor in human osteosarcomas. Exposure to demethylating agents caused reexpression of estrogen receptor, which promoted therapeutic differentiation of these tumors.See related article by Lillo Osuna et al., p. 1054.
Collapse
|
17
|
Estrogen signaling in arcuate Kiss1 neurons suppresses a sex-dependent female circuit promoting dense strong bones. Nat Commun 2019; 10:163. [PMID: 30635563 PMCID: PMC6329772 DOI: 10.1038/s41467-018-08046-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 12/06/2018] [Indexed: 11/16/2022] Open
Abstract
Central estrogen signaling coordinates energy expenditure, reproduction, and in concert with peripheral estrogen impacts skeletal homeostasis in females. Here, we ablate estrogen receptor alpha (ERα) in the medial basal hypothalamus and find a robust bone phenotype only in female mice that results in exceptionally strong trabecular and cortical bones, whose density surpasses other reported mouse models. Stereotaxic guided deletion of ERα in the arcuate nucleus increases bone mass in intact and ovariectomized females, confirming the central role of estrogen signaling in this sex-dependent bone phenotype. Loss of ERα in kisspeptin (Kiss1)-expressing cells is sufficient to recapitulate the bone phenotype, identifying Kiss1 neurons as a critical node in this powerful neuroskeletal circuit. We propose that this newly-identified female brain-to-bone pathway exists as a homeostatic regulator diverting calcium and energy stores from bone building when energetic demands are high. Our work reveals a previously unknown target for treatment of age-related bone disease. Estrogen promotes negative energy balance and preserves skeletal physiology. Here the authors show that loss of estrogen signalling after ablating estrogen receptor alpha (ERa) in specific hypothalamic neuronal populations leads to a marked sex-dependent increase in bone mass in female mice.
Collapse
|
18
|
Perkins MS, Louw-du Toit R, Africander D. Hormone Therapy and Breast Cancer: Emerging Steroid Receptor Mechanisms. J Mol Endocrinol 2018; 61:R133-R160. [PMID: 29899079 DOI: 10.1530/jme-18-0094] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/04/2018] [Accepted: 06/12/2018] [Indexed: 12/31/2022]
Abstract
Although hormone therapy is widely used by millions of women to relieve symptoms of menopause, it has been associated with several side-effects such as coronary heart disease, stroke and increased invasive breast cancer risk. These side-effects have caused many women to seek alternatives to conventional hormone therapy, including the controversial custom-compounded bioidentical hormone therapy suggested to not increase breast cancer risk. Historically estrogens and the estrogen receptor were considered the principal factors promoting breast cancer development and progression, however, a role for other members of the steroid receptor family in breast cancer pathogenesis is now evident, with emerging studies revealing an interplay between some steroid receptors. In this review, we discuss examples of hormone therapy used for the relief of menopausal symptoms, highlighting the distinction between conventional hormone therapy and custom-compounded bioidentical hormone therapy. Moreover, we highlight the fact that not all hormones have been evaluated for an association with increased breast cancer risk. We also summarize the current knowledge regarding the role of steroid receptors in mediating the carcinogenic effects of hormones used in menopausal hormone therapy, with special emphasis on the influence of the interplay or crosstalk between steroid receptors. Unraveling the intertwined nature of steroid hormone receptor signaling pathways in breast cancer biology is of utmost importance, considering that breast cancer is the most prevalent cancer among women worldwide. Moreover, understanding these mechanisms may reveal novel prevention or treatment options, and lead to the development of new hormone therapies that does not cause increased breast cancer risk.
Collapse
Affiliation(s)
- Meghan S Perkins
- Department of Biochemistry, Stellenbosch University, Matieland, South Africa
| | - Renate Louw-du Toit
- Department of Biochemistry, Stellenbosch University, Matieland, South Africa
| | - Donita Africander
- Department of Biochemistry, Stellenbosch University, Matieland, South Africa
| |
Collapse
|
19
|
The Evolution of Estrogen Receptor Signaling in the Progression of Endometriosis to Endometriosis-Associated Ovarian Cancer. Discov Oncol 2018; 9:399-407. [PMID: 30302736 DOI: 10.1007/s12672-018-0350-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/19/2018] [Indexed: 01/01/2023] Open
Abstract
To investigate changes in estrogen receptor alpha (ERα) signaling during progression of endometriosis to endometriosis-associated ovarian cancer (EAOC) as a driver of malignant transformation. We procured tissue samples of normal endometrium, endometriosis (benign, atypical, concurrent with EAOC), and EAOC. We evaluated expression of a 236-gene signature of estrogen signaling. ANOVA and unsupervised clustering were used to identify gene expression profiles across disease states. These profiles were compared to profiles of estrogen regulation in cancer models from the Gene Expression Omnibus (GEO). Gene Set Enrichment Analysis (GSEA) was performed to determine whether gene expression in EAOC was consistent with ERα activity. ANOVA revealed 158 differentially expressed genes (q < 0.05) and unsupervised clustering identified five distinct gene clusters. The estrogen signaling profile of EAOC was not consistent with activated ERα in pre-clinical models. Gene set enrichment analysis did not identify signatures of activated ERα in EAOC but instead identified expression patterns consistent with loss of ERα function and development of endocrine resistance. Gene expression data suggest that ERα signaling becomes inactivated throughout the progression of endometriosis to EAOC. The gene expression pattern in EAOC is more consistent with profiles of endocrine resistance.
Collapse
|
20
|
Song W, Liu QS, Sun Z, Yang X, Zhou Q, Jiang G. Polyfluorinated iodine alkanes regulated distinct breast cancer cell progression through binding with estrogen receptor alpha or beta isoforms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 239:300-307. [PMID: 29665550 DOI: 10.1016/j.envpol.2018.04.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 06/08/2023]
Abstract
Polyfluorinated iodine alkanes (PFIs) are a kind of emerging chemicals with endocrine disrupting effects. Based on the different binding preferences of PFIs to estrogen receptor alpha and beta isoforms (ERα and β), two representative PFIs, dodecafluoro-1,6-diiodohexane (PFHxDI) and tridecafluorohexyl iodide (PFHxI), were selected to evaluate their effects on the proliferation of two kinds of breast cancer cells with different ERα/β expression levels, MCF-7 and T47D. The cell viability assay showed PFHxDI could cause higher cellular toxicity than did PFHxI in both MCF-7 and T47D. MCF-7 with relatively higher ERα/β expression ratio was more vulnerable to the cytotoxic treatments of PFHxI and PFHxDI when compared with T47D cells with relatively lower ERα/β expression ratio. EdU incorporation and cell cycle analysis revealed that, similar to 17β-estrodiol (E2), non-cytotoxic levels of PFHxDI could significantly promote the proliferation of MCF-7 by increasing cell population at S phase (p < 0.01), while T47D proliferation was not influenced by PFHxI exposure due to cell cycle arrest at G2/M phase. The cellular responses caused by estrogenic PFIs were dominantly mediated by their preferential binding affinities for ER isoforms, which would be helpful in the accurate assessment for their potential influences on the breast cancer progression.
Collapse
Affiliation(s)
- Wenting Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; Medical College, Henan Polytechnic University, Jiaozuo, 454000, PR China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhendong Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Institute of Environment and Health, Jianghan University, Wuhan, 430056, PR China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
21
|
Chang BY, Kim DS, Kim HS, Kim SY. Evaluation of estrogenic potential by herbal formula, HPC 03 for in vitro and in vivo. Reproduction 2018; 155:105-115. [PMID: 29326134 DOI: 10.1530/rep-17-0530] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/29/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022]
Abstract
HPC 03 is herbal formula that consists of extracts from Angelica gigas, Cnidium officinale Makino and Cinnamomum cassia Presl. The present study evaluated the estrogenic potential of HPC 03 by using in vitro and in vivo models. The regulatory mechanisms of HPC 03 in estrogen-dependent MCF-7 cells were assessed. HPC 03 induced the proliferation of estrogen receptor-positive MCF-7 cells, and the proliferation was blocked by the addition of the estrogen antagonist tamoxifen. The estrogen receptorα/β luciferase activities were significantly increased by HPC 03 treatment, which also increased the mRNA expression of the estrogen-responsive genes Psen2, Pgr and Ctsd Also, we evaluated the ameliorative effects of HPC 03 on menopausal symptoms in ovariectomized rats. HPC 03 treatment in OVX rats significantly affected the uterine weight, increased the expression of estrogen-responsive genes Pgr and Psen2 in uterus, increased bone mineral density loss in the femur and inhibited body weight increase. Serum E2, collagen type 1 and osteocalcin were significantly increased, while serum LH, FSH and ALP were decreased compared with OVX rats. HPC 03 may be a promising candidate for the treatment of menopause, but further research is necessary to determine whether the observed effects also occur in humans.
Collapse
Affiliation(s)
- Bo Yoon Chang
- Institute of Pharmaceutical Research and DevelopmentCollege of Pharmacy, Wonkwang University, Iksan, Jeonbuk, South Korea
| | - Dae Sung Kim
- Hanpoong Pharm. Co. LtdJeonju-si, Jeonbuk, South Korea
| | - Hye Soo Kim
- Hanpoong Pharm. Co. LtdJeonju-si, Jeonbuk, South Korea
| | - Sung Yeon Kim
- Institute of Pharmaceutical Research and DevelopmentCollege of Pharmacy, Wonkwang University, Iksan, Jeonbuk, South Korea
| |
Collapse
|
22
|
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
|
23
|
Lu S, Sun C, Miao C, Zhao Z. ERβ compensates for the absence of ERα function to promote osteoblast viability by inhibition of SOST signaling. Exp Ther Med 2017; 14:3387-3392. [PMID: 29042923 PMCID: PMC5639354 DOI: 10.3892/etm.2017.5014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 01/26/2017] [Indexed: 01/01/2023] Open
Abstract
Estrogen receptors α and β (ERα and ERβ) serve key functions in bone development and maintenance, and in the metabolism of bone mineral. ERβ and ERα form heterodimers, and ERβ negatively regulates the transactivation of ERα. ERβ also inhibits recruitment of ERα to the estrogen-responsive promoters. However, the relationship of ERα and ERβ in the regulation of osteoblast viability and differentiation remains unclear. The present study aimed to investigate whether ERβ plays a role in balancing ERα activity in osteoblast cells. Downregulation of ERα by short hairpin RNA (shRNA) was found to significantly increase cell cycle arrest at G1 phase (P<0.01). In addition, this effect was found to be significantly enhanced by downregulation of ERβ (P<0.05). Inversely, ERα-knocked down osteoblasts were treated with ERβ agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) to activate ERβ. It was found that activation of ERβ significantly rescued the arrest of cell cycle induced by the downregulation of ERα (P<0.05). Furthermore, downregulation of ERα was found to significantly inhibit cell viability (P<0.01), and knockdown of ERβ was found to have a significant synergic effect with ERα downregulation on the inhibition of cell viability (P<0.01). Treatment with ERβ agonist DPN significantly rescued the effects of downregulation of ERα on cell viability (P<0.01). It was also demonstrated that the synergic effects of ERα and ERβ deletion was via upregulation of SOST gene expression, and the subsequent inhibition of OPG and Runx2 gene expression. Thus, ERβ may serve a function in balancing osteoblast viability and differentiation induced by ERα.
Collapse
Affiliation(s)
- Shijin Lu
- Department of Orthopedics, The Affiliated Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Changying Sun
- Department of Orthopedics, The Affiliated Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Congxiu Miao
- Department of Orthopedics, The Affiliated Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Zhongfu Zhao
- Department of Orthopedics, The Affiliated Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| |
Collapse
|
24
|
Tian K, Qi W, Yan Q, Zhang F, Song D, Zhang H, Lv M. Combined analysis of ChIP-seq and gene microarray datasets identify the E2-mediated genes in ERα-dependent manner in osteosarcoma. Oncol Rep 2017; 38:2335-2342. [PMID: 28849169 DOI: 10.3892/or.2017.5914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/11/2017] [Indexed: 11/05/2022] Open
Abstract
Osteosarcoma is a common bone tumor which is affected by E2, the most representative estrogen. Gene regulation function of E2 is highly dependent on estrogen receptor. The purpose of this study was to explore the gene regulation patterns of E2 through estrogen receptor α (ESR1) in osteosarcoma based on the combined analysis of ChIP-seq and gene microarray. All of the datasets were downloaded from the Gene Expression Omnibus (GEO). Differential expression genes (DEGs) in E2 treated U2OS cells expressing ESR1 (U2OS-ERα) compared with those treated with vehicle were obtained based on R programming software. ESR1-specific binding sites (peaks) in E2 treated U2OS cells were identified through MACS. Overlaps between DEGs and ESR1 target genes which contained peaks in promoters were considered as reliable E2-mediated genes through ESR1 in osteosarcoma. Moreover, we conducted miRNA-Gene regulation analysis for those genes through miRWalk database to identify potential therapeutic targets for the genes. Functional enrichment analysis of DEGs indicated their potential involvement in cancer, and cell activity-related processes. Fifteen overlaps were identified between DEGs and target genes of ESR1, of which 12 were found to be regulated by miRNA. Several known estrogen response genes and novel genes were obtained in this study and they might provide potential therapeutic targets for osteosarcoma.
Collapse
Affiliation(s)
- Kangsong Tian
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Wei Qi
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Qian Yan
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Feng Zhang
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Delei Song
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Haiyang Zhang
- Microscopic Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Ming Lv
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
25
|
Regulation of hepatic Na+/K+-ATPase in obese female and male rats: involvement of ERK1/2, AMPK, and Rho/ROCK. Mol Cell Biochem 2017; 440:77-88. [DOI: 10.1007/s11010-017-3157-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/05/2017] [Indexed: 12/22/2022]
|
26
|
Song W, Zhao L, Sun Z, Yang X, Zhou Q, Jiang G. A novel high throughput screening assay for binding affinities of perfluoroalkyl iodide for estrogen receptor alpha and beta isoforms. Talanta 2017; 175:413-420. [PMID: 28842010 DOI: 10.1016/j.talanta.2017.07.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/17/2017] [Accepted: 07/23/2017] [Indexed: 11/26/2022]
Abstract
Contaminants of emerging concern are continuously increasing, which makes it important to develop high throughput screening techniques for the evaluation of their potential biological effects, especially endocrine disrupting effects, which would directly influence the population dynamics in environment. A novel competitive binding assay based on enzyme fragmentation complementation technology was established to screen the binding affinities of emerging chemicals for estrogen receptor (ER) α or β isoforms. Exogenous compounds could compete with the fragment (ED-ES) of genetically engineered β-galactosidase enzyme (β-gal) for the binding to ERα or β, thus quantitatively altering the formation of enzymatically active β-gal and the hydrolysis of luminescent substrate. According to the monitoring of luminescence curves and the optimization of ERα or β concentrations, it was found that luminescent signals were sustainably emitted for 9h, and 40nM ERα or β in the system would lead to the most sensitive luminescence response. Using 17β-estrodiol (E2) and genistein as the representative estrogenic hormones, their binding affinities for ERα and β were evaluated. The results were consistent with those determined by traditional methods, which confirmed the reliability of this competitive binding assay based on β-gal. Four polyfluorinated iodine alkanes (PFIs) with specific structural characteristics in iodine substitution and carbon chain length were screened, and the results showed diverse binding affinities and different preferences of these chemicals to ERα or β isoforms. The binding affinities of PFIs for ERα were consistent with the result from MVLN transcriptional reporter assay. Overall, the competitive binding assay presented in this study provided a promising alternative to high throughput screening of emerging chemicals with estrogenic effects, which would be important in explanation of their potential toxicological effects and human exposure risks.
Collapse
Affiliation(s)
- Wenting Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; Medical College, Henan Polytechnic University, Jiaozuo 454000, PR China
| | - Lixia Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zhendong Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
27
|
Rietjens IMCM, Louisse J, Beekmann K. The potential health effects of dietary phytoestrogens. Br J Pharmacol 2017; 174:1263-1280. [PMID: 27723080 PMCID: PMC5429336 DOI: 10.1111/bph.13622] [Citation(s) in RCA: 282] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/04/2016] [Accepted: 09/05/2016] [Indexed: 12/14/2022] Open
Abstract
Phytoestrogens are plant-derived dietary compounds with structural similarity to 17-β-oestradiol (E2), the primary female sex hormone. This structural similarity to E2 enables phytoestrogens to cause (anti)oestrogenic effects by binding to the oestrogen receptors. The aim of the present review is to present a state-of-the-art overview of the potential health effects of dietary phytoestrogens. Various beneficial health effects have been ascribed to phytoestrogens, such as a lowered risk of menopausal symptoms like hot flushes and osteoporosis, lowered risks of cardiovascular disease, obesity, metabolic syndrome and type 2 diabetes, brain function disorders, breast cancer, prostate cancer, bowel cancer and other cancers. In contrast to these beneficial health claims, the (anti)oestrogenic properties of phytoestrogens have also raised concerns since they might act as endocrine disruptors, indicating a potential to cause adverse health effects. The literature overview presented in this paper illustrates that several potential health benefits of phytoestrogens have been reported but that, given the data on potential adverse health effects, the current evidence on these beneficial health effects is not so obvious that they clearly outweigh the possible health risks. Furthermore, the data currently available are not sufficient to support a more refined (semi) quantitative risk-benefit analysis. This implies that a definite conclusion on possible beneficial health effects of phytoestrogens cannot be made. LINKED ARTICLES This article is part of a themed section on Principles of Pharmacological Research of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.11/issuetoc.
Collapse
Affiliation(s)
| | - Jochem Louisse
- Division of ToxicologyWageningen UniversityWageningenThe Netherlands
| | - Karsten Beekmann
- Division of ToxicologyWageningen UniversityWageningenThe Netherlands
| |
Collapse
|
28
|
Li H, He Y, Hao P, Liu P. Identification of characteristic gene modules of osteosarcoma using bioinformatics analysis indicates the possible molecular pathogenesis. Mol Med Rep 2017; 15:2113-2119. [PMID: 28259906 PMCID: PMC5364958 DOI: 10.3892/mmr.2017.6245] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 12/06/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the possible pathogenesis of osteosarcoma using bioinformatics analysis to examine gene‑gene interactions. A total of three datasets associated with osteosarcoma were downloaded from the Gene Expression Omnibus. The differentially expressed genes (DEGs) were identified using the significance analysis of microarrays method, which then were subjected to the Human Protein Reference Database to identify the protein‑protein interaction (PPI) pairs and to construct a PPI network of the DEGs. Subsequent multilevel community analysis was applied to mine the modules in the network, followed by screening of the differential expression module using the GlobalAncova package. The genes in the differential expression modules were verified in the valid datasets. The verified genes underwent functional and pathway enrichment analysis. A total of 616 DEGs were selected to construct the PPI network, which included 5,808 osteosarcoma‑specific interaction pairs and 8,012 normal‑specific pairs. Tumor protein p53 (TP53), mitogen-activated protein kinase 1 (MAPK1) and estrogen receptor 1 (ESR1) were identified the most important osteosarcoma‑associated genes, with the highest levels of topological properties. Neurogenic locus notch homolog protein 3 (NOTCH3) and caspase 1 (CASP1) were identified as the osteosarcoma‑specific interaction pairs. Among all 23 mined modules, three were identified as differential expression modules, which were verified in the other two datasets. The genes in these modules were predominantly enriched in the FGFR, MAPK and Notch signaling pathways. Therefore, TP53, MAPK1, ESR1, NOTCH3 and CASP1 may be important in the development of osteosarcoma, and provides valuable clues to investigate the pathogenesis of osteosarcoma using the three differential expression modules.
Collapse
Affiliation(s)
- Hongmin Li
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
- Cancer Center, Affiliated Medical School, University of Electronic Science and Technology, Chengdu, Sichuan 610051, P.R. China
| | - Yangke He
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
- Cancer Center, Affiliated Medical School, University of Electronic Science and Technology, Chengdu, Sichuan 610051, P.R. China
| | - Peng Hao
- Department of Orthopedics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
- Department of Orthopedics, Affiliated Medical School, University of Electronic Science and Technology, Chengdu, Sichuan 610051, P.R. China
| | - Pan Liu
- Department of Orthopedics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
- Department of Orthopedics, Affiliated Medical School, University of Electronic Science and Technology, Chengdu, Sichuan 610051, P.R. China
| |
Collapse
|
29
|
Lemler DJ, Lynch ML, Tesfay L, Deng Z, Paul BT, Wang X, Hegde P, Manz DH, Torti SV, Torti FM. DCYTB is a predictor of outcome in breast cancer that functions via iron-independent mechanisms. Breast Cancer Res 2017; 19:25. [PMID: 28270217 PMCID: PMC5341190 DOI: 10.1186/s13058-017-0814-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 02/09/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Duodenal cytochrome b (DCYTB) is a ferrireductase that functions together with divalent metal transporter 1 (DMT1) to mediate dietary iron reduction and uptake in the duodenum. DCYTB is also a member of a 16-gene iron regulatory gene signature (IRGS) that predicts metastasis-free survival in breast cancer patients. To better understand the relationship between DCYTB and breast cancer, we explored in detail the prognostic significance and molecular function of DCYTB in breast cancer. METHODS The prognostic significance of DCYTB expression was evaluated using publicly available microarray data. Signaling Pathway Impact Analysis (SPIA) of microarray data was used to identify potential novel functions of DCYTB. The role of DCYTB was assessed using immunohistochemistry and measurements of iron uptake, iron metabolism, and FAK signaling. RESULTS High DCYTB expression was associated with prolonged survival in two large independent cohorts, together totaling 1610 patients (cohort #1, p = 1.6e-11, n = 741; cohort #2, p = 1.2e-05, n = 869; log-rank test) as well as in the Gene expression-based Outcome for Breast cancer Online (GOBO) cohort (p < 1.0e-05, n = 1379). High DCYTB expression was also associated with increased survival in homogeneously treated groups of patients who received either tamoxifen or chemotherapy. Immunohistochemistry revealed that DCYTB is localized on the plasma membrane of breast epithelial cells, and that expression is dramatically reduced in high-grade tumors. Surprisingly, neither overexpression nor knockdown of DCYTB affected levels of ferritin H, transferrin receptor, labile iron or total cellular iron in breast cancer cells. Because SPIA pathway analysis of patient microarray data revealed an association between DCYTB and the focal adhesion pathway, we examined the influence of DCYTB on FAK activation in breast cancer cells. These experiments reveal that DCYTB reduces adhesion and activation of focal adhesion kinase (FAK) and its adapter protein paxillin. CONCLUSIONS DCYTB is an important predictor of outcome and is associated with response to therapy in breast cancer patients. DCYTB does not affect intracellular iron in breast cancer cells. Instead, DCYTB may retard cancer progression by reducing activation of FAK, a kinase that plays a central role in tumor cell adhesion and metastasis.
Collapse
Affiliation(s)
- David J. Lemler
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
- Present address: Department of Molecular Biomedical Sciences, North Carolina State University, CVM Research Building 474, Raleigh, NC 27695 USA
| | - Miranda L. Lynch
- Center for Quantitative Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
- Present address: Statistical Sciences Group CCS-6, Los Alamos National Laboratory, Los Alamos, NM 87545 USA
| | - Lia Tesfay
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Zhiyong Deng
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Bibbin T. Paul
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Xiaohong Wang
- Department of Pathology, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Poornima Hegde
- Department of Pathology, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - David H. Manz
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
- School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Suzy V. Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Frank M. Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| |
Collapse
|
30
|
Liu Y, Hou Y, Ma L, Sun C, Pan J, Yang Y, Zhou H, Zhang J. Regulation of semaphorin 4D expression and cell proliferation of ovarian cancer by ERalpha and ERbeta. ACTA ACUST UNITED AC 2017; 50:e6057. [PMID: 28225892 PMCID: PMC5333722 DOI: 10.1590/1414-431x20166057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/13/2016] [Indexed: 12/01/2022]
Abstract
Ovarian cancer is one of the most common malignancies in women. Semaphorin 4D (sema 4D) is involved in the progress of multiple cancers. In the presence of estrogen-like ligands, estrogen receptors (ERα and ERβ) participate in the progress of breast and ovarian cancers by transcriptional regulation. The aim of the study was to investigate the role of sema 4D and elucidate the regulatory pattern of ERα and ERβ on sema 4D expression in ovarian cancers. Sema 4D levels were up-regulated in ovarian cancer SKOV-3 cells. Patients with malignant ovarian cancers had significantly higher sema 4D levels than controls, suggesting an oncogene role of sema 4D in ovarian cancer. ERα expressions were up-regulated in SKOV-3 cells compared with normal ovarian IOSE80 epithelial cells. Conversely, down-regulation of ERβ was observed in SKOV-3 cells. Forced over-expression of ERα and ERβ in SKOV-3 cells was manipulated to establish ERα+ and ERβ+ SKOV-3 cell lines. Incubation of ERα+ SKOV-3 cells with ERs agonist 17β-estradiol (E2) significantly enhanced sema 4D expression and rate of cell proliferation. Incubated with E2, ERβ+ SKOV-3 cells showed lower sema 4D expression and cell proliferation. Blocking ERα and ERβ activities with ICI182-780 inhibitor, sema 4D expressions and cell proliferation of ERα+ and ERβ+ SKOV-3 cells were recovered to control levels. Taken together, the data showed that sema 4D expression was positively correlated with the progress of ovarian cancer. ERα positively regulated sema 4D expression and accelerated cell proliferation. ERβ negatively regulated sema 4D expression and inhibited cell multiplication.
Collapse
Affiliation(s)
- Y Liu
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Y Hou
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - L Ma
- Department of Reproduction and Genetics, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - C Sun
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - J Pan
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Y Yang
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - H Zhou
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - J Zhang
- Department of General Surgery, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
31
|
Szwejser E, Maciuszek M, Casanova-Nakayama A, Segner H, Verburg-van Kemenade BML, Chadzinska M. A role for multiple estrogen receptors in immune regulation of common carp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:61-72. [PMID: 27062969 DOI: 10.1016/j.dci.2016.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/05/2016] [Accepted: 04/05/2016] [Indexed: 05/02/2023]
Abstract
Estrogens are important for bi-directional neuroendocrine-immune interaction. They act via nuclear estrogen receptors (ERα and ERβ) and/or G-protein coupled receptor - GPR30. We found expression of ERα, ERβ and GPR30 in carp lymphoid tissues and head kidney monocytes/macrophages, neutrophils and lymphocytes. Interestingly, ERβ is also expressed in some head kidney lymphocytes but not in naive PBLs. Immune stimulation altered the cell type specific profile of expression of these receptors, which depends on both activation and maturation stage. This implies direct leukocyte responsiveness to estrogen stimulation and therefore in vitro effects of 17β-estradiol (E2) on reactive oxygen species (ROS) production in monocytes/macrophages were determined. Short-time incubation with E2 increased ROS production in PMA-stimulated cells. Results comply with mediation by GPR30, partially functioning via phosphoinositide 3-kinase activation. These results furthermore demonstrate that neuroendocrine-immune communication via estrogen receptors is evolutionary conserved.
Collapse
Affiliation(s)
- Ewa Szwejser
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland
| | - Magdalena Maciuszek
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland
| | - Ayako Casanova-Nakayama
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland
| | - Helmut Segner
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland
| | - B M Lidy Verburg-van Kemenade
- Cell Biology and Immunology Group, Dept of Animal Sciences, Wageningen University, P.O. Box 338, 6700 AH Wageningen, The Netherlands
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland.
| |
Collapse
|
32
|
Yuan B, Cheng L, Gupta K, Chiang HC, Gupta HB, Sareddy GR, Wang D, Lathrop K, Elledge R, Wang P, McHardy S, Vadlamudi R, Curiel TJ, Hu Y, Ye Q, Li R. Tyrosine phosphorylation regulates ERβ ubiquitination, protein turnover, and inhibition of breast cancer. Oncotarget 2016; 7:42585-42597. [PMID: 27323858 PMCID: PMC5173158 DOI: 10.18632/oncotarget.10018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 05/23/2016] [Indexed: 12/31/2022] Open
Abstract
Unlike estrogen receptor α (ERα) that predominantly promotes hormone-dependent breast tumor growth, ERβ exhibits antitumor effects in a variety of cancer types. We recently identified a phosphotyrosine residue in ERβ, but not ERα, that dictates ERβ transcriptional activity and antitumor function. We show here that this ER isotype-specific phosphotyrosine switch is important for regulating ERβ activity in cell proliferation, migration, and invasion. At the mechanistic level, phosphorylated ERβ, which recruits transcriptional coactivator p300, is in turn targeted by p300 for ubiquitination and proteasome-dependent protein turnover. Furthermore, ERβ-specific agonists such as S-equol enhance ERβ phosphorylation, suggesting a crosstalk between ligand- and posttranslational modification-dependent ERβ activation. Inhibition of xenograft tumor growth by S-equol is associated with reduced tumor Ki-67 expression and elevated ERβ tyrosine phosphorylation. Taken together, our data support the notion that phosphotyrosine-dependent ERβ signaling is an attractive target for anticancer treatment.
Collapse
Affiliation(s)
- Bin Yuan
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Liaoning, China
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Long Cheng
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Kshama Gupta
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Huai-Chin Chiang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Harshita B. Gupta
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Degeng Wang
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kate Lathrop
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Richard Elledge
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Pei Wang
- Department of Cellular and Structural Biology Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Stanton McHardy
- Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX, USA
| | - Ratna Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tyler J. Curiel
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yanfen Hu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Liaoning, China
| | - Rong Li
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
33
|
Lahsaee S, Corkery DP, Anthes LE, Holly A, Dellaire G. Estrogen receptor alpha (ESR1)-signaling regulates the expression of the taxane-response biomarker PRP4K. Exp Cell Res 2015; 340:125-31. [PMID: 26712520 DOI: 10.1016/j.yexcr.2015.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/12/2015] [Accepted: 12/16/2015] [Indexed: 10/22/2022]
Abstract
The pre-mRNA splicing factor 4 kinase PRP4K (PRPF4B), is an essential kinase that is a component of the U5 snRNP and functions in spliceosome assembly. We demonstrated that PRP4K is a novel biological marker for taxane response in ovarian cancer patients and reduced levels of PRP4K correlate with intrinsic and acquired taxane resistance in both breast and ovarian cancer. Breast cancer treatments are chosen based on hormone and growth factor receptor status, with HER2 (ERBB2) positive breast cancer patients receiving anti-HER2 agents and taxanes and estrogen receptor alpha (ESR1) positive (ER+) breast cancer patients receiving anti-estrogen therapies such as tamoxifen. Here we demonstrate that PRP4K is expressed in the normal mammary duct epithelial cells of the mouse, and that estrogen induces PRP4K gene and protein expression in ER+ human MCF7 breast cancer cells. Estrogen acts through ESR1 to regulate PRP4K expression, as over-expression of ESR1 in the ER-negative MDA-MB-231 breast cancer cell line increased the expression of this kinase, and knock-down of ESR1 in ER+ T47D breast cancer cells reduced PRP4K levels. Furthermore, treatment with 4-hydroxytamoxifen (4-OHT) resulted in a dose-dependent decrease in PRP4K protein expression in MCF7 cells. Consistent with our previous studies identifying PRP4K as a taxane-response biomarker, reduced PRP4K expression in 4-OHT-treated cells correlated with reduced sensitivity to paclitaxel. Thus, PRP4K is novel estrogen regulated kinase, and its levels can be reduced by 4-OHT in ER+ breast cancer cells altering their response to taxanes.
Collapse
Affiliation(s)
- Sara Lahsaee
- Departments of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Dale P Corkery
- Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Livia E Anthes
- Departments of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alice Holly
- Departments of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Graham Dellaire
- Departments of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada; Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
34
|
Kim MH, Kim HM, Jeong HJ. Estrogen-like osteoprotective effects of glycine in in vitro and in vivo models of menopause. Amino Acids 2015; 48:791-800. [PMID: 26563333 DOI: 10.1007/s00726-015-2127-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/04/2015] [Indexed: 01/31/2023]
Abstract
Recently, the placenta mesotherapy has been widely used to treat menopause. Placenta contains amino acids, peptides, minerals, and estrogen. Here, we investigated the estrogen-like osteoprotective effects of glycine (a main ingredient of placenta) in in vitro and in vivo models of menopause. We assessed the effect of glycine on MG-63 osteoblast cell line, MCF-7 estrogen-dependent cell line, and ovariectomized (OVX) mice. Glycine significantly increased the MG-63 cell proliferation in a dose-dependent manner. Activity of alkaline phosphatase (ALP) and phosphorylation of extracellular-signal-regulated kinase were increased by glycine in MG-63 cells. Glycine also increased the BrdU-incorporation and Ki-67 mRNA expression in MCF-7 cells. Glycine induced the up-regulation of estrogen receptor-β mRNA expression and estrogen-response element-luciferase activity in MG-63 and MCF-7 cells. In OVX mice, glycine was administered orally at a daily dose of 10 mg/kg per day for 8 weeks. Glycine resulted in the greatest decrease in weight gain caused by ovariectomy. Meanwhile, vaginal weight reduced by ovariectomy was increased by glycine. Glycine significantly increased the ALP activity in OVX mice. MicroCT-analysis showed that glycine significantly enhanced bone mineral density, trabecular number, and connectivity density in OVX mice. Moreover, glycine significantly increased the serum 17β-estradiol levels reduced by ovariectomy. Glycine has an estrogen-like osteoprotective effect in menopause models. Therefore, we suggest that glycine may be useful for the treatment of menopause.
Collapse
Affiliation(s)
- Min-Ho Kim
- Department of Computer Aided Mechanical Engineering, Sohae College, Gunsan, Jeonbuk, 573-717, Republic of Korea
| | - Hyung-Min Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Hyun-Ja Jeong
- Department of Food Technology and Biochip Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do, 336-795, Republic of Korea.
| |
Collapse
|
35
|
Shi L, Lin Q, Su B. Estrogen regulation of microcephaly genes and evolution of brain sexual dimorphism in primates. BMC Evol Biol 2015; 15:127. [PMID: 26123139 PMCID: PMC4487212 DOI: 10.1186/s12862-015-0398-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/29/2015] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Sexual dimorphism in brain size is common among primates, including humans, apes and some Old World monkeys. In these species, the brain size of males is generally larger than that of females. Curiously, this dimorphism has persisted over the course of primate evolution and human origin, but there is no explanation for the underlying genetic controls that have maintained this disparity in brain size. RESULTS In the present study, we tested the effect of the female hormone (estradiol) on seven genes known to be related to brain size in both humans and nonhuman primates, and we identified half estrogen responsive elements (half EREs) in the promoter regions of four genes (MCPH1, ASPM, CDK5RAP2 and WDR62). Likewise, at sequence level, it appears that these half EREs are generally conserved across primates. Later testing via a reporter gene assay and cell-based endogenous expression measurement revealed that estradiol could significantly suppress the expression of the four affected genes involved in brain size. More intriguingly, when the half EREs were deleted from the promoters, the suppression effect disappeared, suggesting that the half EREs mediate the regulation of estradiol on the brain size genes. We next replicated these experiments using promoter sequences from chimpanzees and rhesus macaques, and observed a similar suppressive effect of estradiol on gene expression, suggesting that this mechanism is conserved among primate species that exhibit brain size dimorphism. CONCLUSIONS Brain size dimorphism among certain primates, including humans, is likely regulated by estrogen through its sex-dependent suppression of brain size genes during development.
Collapse
Affiliation(s)
- Lei Shi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 East Jiao-Chang Road, Kunming, 650223, Yunnan, PR China. .,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650000, China.
| | - Qiang Lin
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 East Jiao-Chang Road, Kunming, 650223, Yunnan, PR China. .,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650000, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China.
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 East Jiao-Chang Road, Kunming, 650223, Yunnan, PR China. .,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650000, China.
| |
Collapse
|
36
|
H19 lncRNA mediates 17β-estradiol-induced cell proliferation in MCF-7 breast cancer cells. Oncol Rep 2015; 33:3045-52. [PMID: 25846769 DOI: 10.3892/or.2015.3899] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/10/2015] [Indexed: 11/05/2022] Open
|
37
|
Reese JM, Suman VJ, Subramaniam M, Wu X, Negron V, Gingery A, Pitel KS, Shah SS, Cunliffe HE, McCullough AE, Pockaj BA, Couch FJ, Olson JE, Reynolds C, Lingle WL, Spelsberg TC, Goetz MP, Ingle JN, Hawse JR. ERβ1: characterization, prognosis, and evaluation of treatment strategies in ERα-positive and -negative breast cancer. BMC Cancer 2014; 14:749. [PMID: 25288324 PMCID: PMC4196114 DOI: 10.1186/1471-2407-14-749] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/25/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The role and clinical value of ERβ1 expression is controversial and recent data demonstrates that many ERβ antibodies are insensitive and/or non-specific. Therefore, we sought to comprehensively characterize ERβ1 expression across all sub-types of breast cancer using a validated antibody and determine the roles of this receptor in mediating response to multiple forms of endocrine therapy both in the presence and absence of ERα expression. METHODS Nuclear and cytoplasmic expression patterns of ERβ1 were analyzed in three patient cohorts, including a retrospective analysis of a prospective adjuvant tamoxifen study and a triple negative breast cancer cohort. To investigate the utility of therapeutically targeting ERβ1, we generated multiple ERβ1 expressing cell model systems and determined their proliferative responses following anti-estrogenic or ERβ-specific agonist exposure. RESULTS Nuclear ERβ1 was shown to be expressed across all major sub-types of breast cancer, including 25% of triple negative breast cancers and 33% of ER-positive tumors, and was associated with significantly improved outcomes in ERα-positive tamoxifen-treated patients. In agreement with these observations, ERβ1 expression sensitized ERα-positive breast cancer cells to the anti-cancer effects of selective estrogen receptor modulators (SERMs). However, in the absence of ERα expression, ERβ-specific agonists potently inhibited cell proliferation rates while anti-estrogenic therapies were ineffective. CONCLUSIONS Using a validated antibody, we have confirmed that nuclear ERβ1 expression is commonly present in breast cancer and is prognostic in tamoxifen-treated patients. Using multiple breast cancer cell lines, ERβ appears to be a novel therapeutic target. However, the efficacy of SERMs and ERβ-specific agonists differ as a function of ERα expression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 16-01B Guggenheim Building, 200 First St, SW, Rochester, MN 55905, USA.
| |
Collapse
|
38
|
Evers NM, van den Berg JHJ, Wang S, Melchers D, Houtman R, de Haan LHJ, Ederveen AGH, Groten JP, Rietjens IMCM. Cell proliferation and modulation of interaction of estrogen receptors with coregulators induced by ERα and ERβ agonists. J Steroid Biochem Mol Biol 2014; 143:376-85. [PMID: 24923734 DOI: 10.1016/j.jsbmb.2014.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 05/27/2014] [Accepted: 06/06/2014] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to investigate modulation of the interaction of the ERα and ERβ with coregulators in the ligand responses induced by estrogenic compounds. To this end, selective ERα and ERβ agonists were characterized for intrinsic relative potency reflected by EC50 and maximal efficacy towards ERα and ERβ mediated response in ER selective reporter gene assays, and subsequently tested for induction of cell proliferation in T47D-ERβ cells with variable ERα/ERβ ratio, and finally for ligand dependent modulation of the interaction of ERα and ERβ with coregulators using the MARCoNI assay, with 154 unique nuclear receptor coregulator peptides derived from 66 different coregulators. Results obtained reveal an important influence of the ERα/ERβ ratio and receptor selectivity of the compounds tested on induction of cell proliferation. ERα agonists activate cell proliferation whereas ERβ suppresses ERα mediated cell proliferation. The responses in the MARCoNI assay reveal that upon ERα or ERβ activation by a specific agonist, the modulation of the interaction of the ERs with coregulators is very similar indicating only a limited number of differences upon ERα or ERβ activation by a specific ligand. Differences in the modulation of the interaction of the ERs with coregulators between the different agonists were more pronounced. Based on ligand dependent differences in the modulation of the interaction of the ERs with coregulators, the MARCoNI assay was shown to be able to classify the ER agonists discriminating between different agonists for the same receptor, a characteristic not defined by the ER selective reporter gene or proliferation assays. It is concluded that the ultimate effect of the model compounds on proliferation of estrogen responsive cells depends on the intrinsic relative potency of the agonist towards ERα and ERβ and the cellular ERα/ERβ ratio whereas differences in the modulation of the interaction of the ERα and ERβ with coregulators contribute to the ligand dependent responses induced by estrogenic compounds.
Collapse
Affiliation(s)
- Nynke M Evers
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands.
| | | | - Si Wang
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands
| | - Diana Melchers
- PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, the Netherlands
| | - René Houtman
- PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, the Netherlands
| | - Laura H J de Haan
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands
| | - Antwan G H Ederveen
- Pharmacokinetics Pharmacodynamics & Drug Metabolism, MSD, P.O. Box 20, 5340 BH Oss, the Netherlands
| | - John P Groten
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands; PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, the Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands
| |
Collapse
|
39
|
Evers NM, Wang S, van den Berg JHJ, Houtman R, Melchers D, de Haan LHJ, Ederveen AGH, Groten JP, Rietjens IMCM. Identification of coregulators influenced by estrogen receptor subtype specific binding of the ER antagonists 4-hydroxytamoxifen and fulvestrant. Chem Biol Interact 2014; 220:222-30. [PMID: 25014417 DOI: 10.1016/j.cbi.2014.06.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/07/2014] [Accepted: 06/19/2014] [Indexed: 11/18/2022]
Abstract
The aim of the present study was to investigate modulation of the interaction of ERα and ERβ with coregulators in the ligand dependent responses induced by the ER antagonistic compounds 4OHT and fulvestrant. Comparison with the modulation index (MI) profiles for the ER agonist estradiol (E2) will elucidate whether differences in the (ant)agonist dependent interaction of ERα and ERβ with coregulators expressed in MI profiles contribute to the differences in (ant)agonist responses. To this end, the selected ER antagonistic compounds were first characterized for intrinsic relative potency and efficacy towards ERα and ERβ using ER selective U2OS reporter gene assays, and subsequently tested for ligand dependent modulation of the interaction of ERα and ERβ with coregulators using the MARCoNI assay. Results obtained indicate a preference of 4OHT to antagonize ERβ and find fulvestrant to be less ER specific. MARCoNI assay responses reveal that ERα and ERβ mediated interaction with coregulators expressed in MI profiles are similar for 4OHT and fulvestrant and generally opposite to the MI profile of the ER agonist E2. Hierarchical clustering based on the MI profiles appeared able to clearly discriminate the two compounds with ER antagonistic properties from the ER agonist E2. Taken together the data reveal that modulation of the interaction of ERs with coregulators discriminates ER agonists from antagonists but does not discriminate between the less specific ER antagonist fulvestrant and the preferential ERβ antagonistic compound 4OHT. It is concluded that differences in modulation of the interaction of ERα and ERβ with coregulators contribute to the differences in ligand dependent responses induced by ER agonists and ER antagonists but the importance of the subtle differences in modulation of the interaction of ERs with coregulators between the ER antagonistic compounds 4OHT and fulvestrant for the ultimate biological effect remains to be established.
Collapse
Affiliation(s)
- Nynke M Evers
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands.
| | - Si Wang
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands
| | | | - René Houtman
- PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, The Netherlands
| | - Diana Melchers
- PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, The Netherlands
| | - Laura H J de Haan
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands
| | - Antwan G H Ederveen
- Pharmacokinetics Pharmacodynamics & Drug Metabolism, MSD, P.O. Box 20, 5340 BH Oss, The Netherlands
| | - John P Groten
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands; PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands
| |
Collapse
|
40
|
Hodgkinson KM, Vanderhyden BC. Consideration of GREB1 as a potential therapeutic target for hormone-responsive or endocrine-resistant cancers. Expert Opin Ther Targets 2014; 18:1065-76. [PMID: 24998469 DOI: 10.1517/14728222.2014.936382] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Steroid hormones increase the incidence and promote the progression of many types of cancer. Exogenous estrogens increase the risk of developing breast, ovarian and endometrial cancer and many breast cancers initially respond to estrogen deprivation. Although steroid hormone signaling has been extensively studied, the mechanisms of hormone-stimulated cancer growth have not yet been fully elucidated, limiting opportunities for novel approaches to therapeutic intervention. AREAS COVERED This review examines growing evidence for the important role played by the steroid hormone-induced gene called GREB1, or growth regulation by estrogen in breast cancer 1. GREB1 is a critical mediator of both the estrogen-stimulated proliferation of breast cancer cells and the androgen-stimulated proliferation of prostate cancer cells. EXPERT OPINION Although its exact function in the cascade of hormone action remains unclear, the ability of GREB1 to modulate tumor progression in models of breast, ovarian and prostate cancer renders this gene an excellent candidate for further consideration as a potential therapeutic target. Research examining the mechanism of GREB1 action will help to elucidate its role in proliferation and its potential contribution to endocrine resistance and will determine whether GREB1 interference may have therapeutic efficacy.
Collapse
Affiliation(s)
- Kendra M Hodgkinson
- Ottawa Hospital Research Institute, Centre for Cancer Therapeutics , 501 Smyth Road, 3rd Floor, Box 926, Ottawa, Ontario K1H 8L6 , Canada
| | | |
Collapse
|
41
|
Ma YL, Qin P, Feng DY, Li Y, Zhang LX, Liu ZY, Yin AQ, Tang WH, Dong HL, Meng LZ, Hou WG, Xiong LZ. Estrogen regulates the expression of Ndrg2 in astrocytes. Brain Res 2014; 1569:1-8. [PMID: 24796879 DOI: 10.1016/j.brainres.2014.04.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 04/17/2014] [Accepted: 04/26/2014] [Indexed: 01/29/2023]
Abstract
N-myc downstream-regulated gene 2 (Ndrg2) is a newly identified molecule that is mainly expressed in astrocytes within the central nervous system (CNS) and is involved in the proliferation and activation of astrocytes. 17β-estradiol (E2) is one of the most important circulating hormones, and in the CNS, astrocytes are a target and potential mediator of the action of E2. Our most recent study found that DPN, an estrogen receptor (ER) β-specific agonist, activated the Ndrg2 promoter and elevated endogenous NDRG2 protein expression in MCF7, HSG and T-47D cells. However, whether E2 regulates Ndrg2 expression in astrocytes remains unknown. Here, we conducted both in vivo and in vitro experiments and found that ERβ co-localized with NDRG2 in astrocytes. Furthermore, in primary cultured astrocytes, we demonstrated that E2 up-regulated Ndrg2 mRNA and protein expression in a dose- and time-dependent manner and that the ERβ agonist DPN but not the ERα agonist PPT up-regulated Ndrg2 expression. In vivo, we found that in the hippocampus of adult ovariectomized (OVX) female mice, Ndrg2 mRNA and protein expression were significantly decreased compared with those in normal adult female mice. After the OVX mice received continuous subcutaneous injections of 50μg/kg E2, 100μg/kg E2 or the ERβ agonist DPN for 10 days, the Ndrg2 expression significantly increased compared with that of the OVX mice. Our results indicate that E2 may affect astrocytes by regulating Ndrg2 expression.
Collapse
Affiliation(s)
- Yu-Long Ma
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China
| | - Pei Qin
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China
| | - Da-Yun Feng
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi׳an 710032, China
| | - Yan Li
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China; Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi׳an 710032, China
| | - Li-Xia Zhang
- Department of Ophthalmology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China
| | - Zhao-Yu Liu
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China
| | - An-Qi Yin
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China
| | - Wen-Hong Tang
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China
| | - Hai-Long Dong
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China
| | - Ling-Zhong Meng
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco 94143-0648, United States
| | - Wu-Gang Hou
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China.
| | - Li-Ze Xiong
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi׳an 710032, China.
| |
Collapse
|
42
|
Flos lonicerae extracts and chlorogenic acid protect human umbilical vein endothelial cells from the toxic damage of perfluorooctane sulphonate. Inflammation 2014; 36:767-79. [PMID: 23392856 DOI: 10.1007/s10753-013-9603-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Chlorogenic acid (CGA), one of the most common phenolic acids, is found in many food and traditional Chinese herbs. Various bioactivities of CGA are studied. However, little is known about these properties of Flos Lonicerae extracts, and the difference in the effect between Flos Lonicerae extracts and CGA has not been reported. CGA was identified in Flos Lonicerae extracts by HPLC and determined qualitatively by quadrupole ion trap mass spectrometry. In this study, we evaluated the effect of Flos Lonicerae extracts and CGA on inflammatory-related gene expression, adhesion molecule expression and reactive oxygen species (ROS) production in perfluorooctane sulphonate (PFOS)-treated human umbilical vein endothelial cells (HUVECs). The suppression of transcription of IL-1β, IL-6, COX-2, and P-Selectin genes with Flos Lonicerae extracts was greater than that of CGA in PFOS-treated HUVECs, while the degree of suppression on PFOS-induced expression of NOS3 and ICAM-1 was greater for CGA. Furthermore, the suppressive effect of Flos Lonicerae extracts on adhesion of monocytes onto PFOS-induced HUVECs was greater than that of CGA. In addition, Flos Lonicerae extracts and CGA were highly effective in reducing ROS although their effects were almost comparable. So, Flos Lonicerae extracts exhibited antioxidant activity and CGA was a major contributor to this activity. These results suggest that Flos Lonicerae extracts could be useful to prevent PFOS-mediated inflammatory diseases.
Collapse
|
43
|
Marciano DP, Chang MR, Corzo CA, Goswami D, Lam VQ, Pascal BD, Griffin PR. The therapeutic potential of nuclear receptor modulators for treatment of metabolic disorders: PPARγ, RORs, and Rev-erbs. Cell Metab 2014; 19:193-208. [PMID: 24440037 DOI: 10.1016/j.cmet.2013.12.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nuclear receptors (NRs) play central roles in metabolic syndrome, making them attractive drug targets despite the challenge of achieving functional selectivity. For instance, members of the thiazolidinedione class of insulin sensitizers offer robust efficacy but have been limited due to adverse effects linked to activation of genes not involved in insulin sensitization. Studies reviewed here provide strategies for targeting subsets of PPARγ target genes, enabling development of next-generation modulators with improved therapeutic index. Additionally, emerging evidence suggests that targeting the NRs ROR and Rev-erb holds promise for treating metabolic syndrome based on their involvement in circadian rhythm and metabolism.
Collapse
Affiliation(s)
- David P Marciano
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Mi Ra Chang
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Cesar A Corzo
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Devrishi Goswami
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Vinh Q Lam
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Bruce D Pascal
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA
| | - Patrick R Griffin
- Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
44
|
Liu L, Zhao Y, Xie K, Sun X, Gao Y, Wang Z. Estrogen-induced nongenomic calcium signaling inhibits lipopolysaccharide-stimulated tumor necrosis factor α production in macrophages. PLoS One 2013; 8:e83072. [PMID: 24376635 PMCID: PMC3871562 DOI: 10.1371/journal.pone.0083072] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 11/07/2013] [Indexed: 12/21/2022] Open
Abstract
Estrogen is traditionally thought to exert genomic actions through members of the nuclear receptor family. Here, we investigated the rapid nongenomic effects of 17β-estradiol (E2) on tumor necrosis factor α (TNF-α) production following lipopolysaccharide (LPS) stimulation in mouse bone marrow-derived macrophages (BMMs). We found that LPS induced TNF-α production in BMMs via phosphorylation of p38 mitogen-activated protein kinase (MAPK). E2 itself did not affect the MAPK pathway, although it attenuated LPS-induced TNF-α production through suppression of p38 MAPK activation. Recently, G protein-coupled receptor 30 (GPR30) was suggested to be a membrane estrogen receptor (mER) that can mediate nongenomic estradiol signaling. We found that BMMs expressed both intracellular estrogen receptors (iER) and mER GPR30. The specific GPR30 antagonist G-15 significantly blocked effects of estradiol on LPS-induced TNF-α production, whereas an iER antagonist did not. Moreover, E2 induced a rapid rise in intracellular free Ca2+ that was due to the influx of extracellular Ca2+ and was not inhibited by an iER antagonist or silencing of iER. Ca2+ influx was also induced by an impermeable E2 conjugated to BSA (E2-BSA), which has been used to investigate the nongenomic effects of estrogen. Consequently, Ca2+, a pivotal factor in E2-stimulated nongenomic action, was identified as the key mediator. The inhibitory effects of E2 on LPS-induced TNF-α production and p38 MAPK phosphorylation were dependent on E2-triggered Ca2+ influx because BAPTA, an intracellular Ca2+ chelator, prevented these effects. Taken together, these data indicate that E2 can down-regulate LPS-induced TNF-α production via blockade of p38 MAPK phosphorylation through the mER-mediated nongenomic Ca2+ signaling pathway in BMMs.
Collapse
Affiliation(s)
- Limin Liu
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Ying Zhao
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Keming Xie
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xiaodong Sun
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yuzhen Gao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, China
- * E-mail: (YZG); (ZFW)
| | - Zufeng Wang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, China
- * E-mail: (YZG); (ZFW)
| |
Collapse
|
45
|
Liu L, Wang Z. Estrogen attenuates lipopolysaccharide-induced nitric oxide production in macrophages partially via the nongenomic pathway. Cell Immunol 2013; 286:53-8. [PMID: 24321566 DOI: 10.1016/j.cellimm.2013.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/29/2013] [Accepted: 11/13/2013] [Indexed: 01/13/2023]
Abstract
Steroid hormones exert genotropic effects through members of the nuclear hormone receptor family. In the present study, we examined the effects of 17β-estradiol (E2) on nitric oxide (NO) production following lipopolysaccharide (LPS) stimulation and investigated the mechanisms in mouse bone marrow-derived macrophages (BMMs). E2 alone did not affect NO production. In contrast, E2 inhibited LPS-induced production of NO in BMMs. Using a cell-impermeable E2 conjugated to BSA (E2-BSA), which has been used to investigate the nongenomic effects of estrogen, we found that the increase in NO production induced by LPS was also attenuated. In addition, the intracellular estrogen receptor blocker, ICI 182780, only partially antagonized the total effects of E2 on LPS-stimulated NO production capacity. E2 also attenuated the LPS activation of p38 mitogen-activated protein kinase (MAPK) but not that of extracellular-regulated protein kinase 1/2 (ERK1/2) and c-Jun NH2-terminal kinase (JNK). This attenuation was not abrogated by ICI 182780. Moreover, the p38 inhibitor, SB 203580, greatly reduced the LPS-induced NO production, and the remaining NO levels were no longer regulated by E2. Additionally, E2-BSA inhibited LPS-mediated changes in p38 MAPK activation to the same extent as E2. Moreover, E2 and E2-BSA inhibited LPS-induced activation of nuclear factor-kappa B (NF-κB) and activator protein 1 (AP-1). This inhibitory effect of E2 was only partially antagonized by ICI 182780. Taken together, these results suggest that E2 has an inhibitory effect on LPS-induced NO production in BMMs through inhibition of p38 MAPK phosphorylation, and blockade of NF-κB and AP-1 activation. These effects are mediated at least in part via a nongenomic pathway.
Collapse
Affiliation(s)
- Limin Liu
- Departments of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Zufeng Wang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
46
|
Wardell JR, Hodgkinson KM, Binder AK, Seymour KA, Korach KS, Vanderhyden BC, Freiman RN. Estrogen responsiveness of the TFIID subunit TAF4B in the normal mouse ovary and in ovarian tumors. Biol Reprod 2013; 89:116. [PMID: 24068106 DOI: 10.1095/biolreprod.113.111336] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Estrogen signaling in the ovary is a fundamental component of normal ovarian function, and evidence also indicates that excessive estrogen is a risk factor for ovarian cancer. We have previously demonstrated that the gonadally enriched TFIID subunit TAF4B, a paralog of the general transcription factor TAF4A, is required for fertility in mice and for the proliferation of ovarian granulosa cells following hormonal stimulation. However, the relationship between TAF4B and estrogen signaling in the normal ovary or during ovarian tumor initiation and progression has yet to be defined. Herein, we show that Taf4b mRNA and TAF4B protein, but not Taf4a mRNA or TAF4A protein, are increased in whole ovaries and granulosa cells of the ovary after exposure to 17beta-estradiol or the synthetic estrogen diethylstilbestrol and that this response occurs within hours after stimulation. Furthermore, this increase occurs via nuclear estrogen receptors both in vivo and in a mouse granulosa cancer cell line, NT-1. We observe a significant increase in Taf4b mRNA in estrogen-supplemented mouse ovarian tumors, which correlates with diminished survival of these mice. These data highlight the novel response of the general transcription factor TAF4B to estrogen in the normal ovary and during ovarian tumor progression in the mouse, suggesting its potential role in regulating actions downstream of estrogen stimulation.
Collapse
Affiliation(s)
- Jennifer R Wardell
- Pathobiology Graduate Program, Brown University, Providence, Rhode Island
| | | | | | | | | | | | | |
Collapse
|
47
|
Williams C, Lin CY. Oestrogen receptors in breast cancer: basic mechanisms and clinical implications. Ecancermedicalscience 2013; 7:370. [PMID: 24222786 PMCID: PMC3816846 DOI: 10.3332/ecancer.2013.370] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Indexed: 12/31/2022] Open
Abstract
Since the discovery of the connection between ovarian hormones and breast cancer, endocrine therapy has been an integral adjuvant treatment for patients with hormone-dependent breast cancers. Oestrogen receptor (ER) plays a central role in mediating the effects of endogenous hormones and therapeutic agents. ER serves as a prognostic marker for responsiveness to endocrine therapy and is targeted either directly by selective oestrogen receptor modulators (SERMs) and pure antagonists or indirectly by aromatase inhibitors (AIs) that block oestrogen production. A significant number of ER-positive patients, however, fail to respond to therapy or develop resistance over time. This review focuses on the current understanding of ER functions and recent advances in genomic technologies and research that have provided a global perspective on hormone and ER activity and led to a number of significant discoveries, including the roles of co-regulatory factors and non-coding RNAs. Mechanistic insights into normal ER functions and therapeutic actions of SERMs and AIs will enable the development of better predictive markers and more effective target mechanisms and ultimately facilitate improvements in disease outcomes and patient survival.
Collapse
Affiliation(s)
- Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204, USA
| | | |
Collapse
|
48
|
Visser K, Mortimer M, Louw A. Cyclopia extracts act as ERα antagonists and ERβ agonists, in vitro and in vivo. PLoS One 2013; 8:e79223. [PMID: 24223909 PMCID: PMC3817056 DOI: 10.1371/journal.pone.0079223] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/20/2013] [Indexed: 12/28/2022] Open
Abstract
Hormone replacement therapy associated risks, and the concomitant reluctance of usage, has instigated the search for new generations of estrogen analogues that would maintain estrogen benefits without associated risks. Furthermore, if these analogues display chemo-preventative properties in breast and endometrial tissues it would be of great value. Both the selective estrogen receptor modulators as well as the selective estrogen receptor subtype modulators have been proposed as estrogen analogues with improved risk profiles. Phytoestrogen containing extracts of Cyclopia, an indigenous South African fynbos plant used to prepare Honeybush tea may serve as a source of new estrogen analogues. In this study three extracts, P104, SM6Met, and cup-of-tea, from two species of Cyclopia, C. genistoides and C. subternata, were evaluated for ER subtype specific agonism and antagonism both in transactivation and transrepression. For transactivation, the Cyclopia extracts displayed ERα antagonism and ERβ agonism when ER subtypes were expressed separately, however, when co-expressed only agonism was uniformly observed. In contrast, for transrepression, this uniform behavior was lost, with some extracts (P104) displaying uniform agonism, while others (SM6Met) displayed antagonism when subtypes were expressed separately and agonism when co-expressed. In addition, breast cancer cell proliferation assays indicate that extracts antagonize cell proliferation in the presence of estrogen at lower concentrations than that required for proliferation. Furthermore, lack of uterine growth and delayed vaginal opening in an immature rat uterotrophic model validates the ERα antagonism of extracts observed in vitro and supports the potential of the Cyclopia extracts as a source of estrogen analogues with a reduced risk profile.
Collapse
Affiliation(s)
- Koch Visser
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
| | - Morné Mortimer
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
| | - Ann Louw
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
- * E-mail:
| |
Collapse
|
49
|
Hsu I, Chuang KL, Slavin S, Da J, Lim WX, Pang ST, O'Brien JH, Yeh S. Suppression of ERβ signaling via ERβ knockout or antagonist protects against bladder cancer development. Carcinogenesis 2013; 35:651-61. [PMID: 24148819 DOI: 10.1093/carcin/bgt348] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epidemiological studies showed that women have a lower bladder cancer (BCa) incidence, yet higher muscle-invasive rates than men, suggesting that estrogen and the estrogen receptors, estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ), may play critical roles in BCa progression. Using in vitro cell lines and an in vivo carcinogen N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced mouse BCa model, we found that ERβ plays a positive role in promoting BCa progression. Knockdown of ERβ with ERβ-shRNA in ERβ-positive human BCa J82, 647v and T24 cell lines led to suppressed cell growth and invasion. Mice lacking ERβ have less cancer incidence with reduced expression of the proliferation marker Ki67 in BBN-induced BCa. Consistently, our results show that non-malignant urothelial cells with ERβ knockdown are more resistant to carcinogen-induced malignant transformation. Mechanism dissection found that targeting ERβ suppressed the expression of minichromosome maintenance complex component 5 (MCM5), a DNA replication licensing factor that is involved in tumor cell growth. Restoring MCM5 expression can partially reverse ERβ knockdown-mediated growth reduction. Supportively, treating cells with the ERβ-specific antagonist, 4-[2-Phenyl-5,7-bis(trifluoromethyl) pyrazolo[1,5-a]pyrimidin-3-yl]phenol (PHTPP), reduced BCa cell growth and invasion, as well as MCM5 expression. Furthermore, we provide the first evidence that BCa burden and mortality can be controlled by PHTPP treatment in the carcinogen-induced BCa model. Together, these results demonstrate that ERβ could play positive roles in promoting BCa progression via MCM5 regulation. Targeting ERβ through ERβ-shRNA, PHTPP or via downstream targets, such as MCM5, could serve as potential therapeutic approaches to battle BCa.
Collapse
|
50
|
Zhang LQ, Zhang XD, Xu J, Wan Y, Qu K, Zhang JY, Wang ZX, Wei JC, Meng FD, Tai MH, Zhou L, Liu C. Potential therapeutic targets for the primary gallbladder carcinoma: estrogen receptors. Asian Pac J Cancer Prev 2013; 14:2185-90. [PMID: 23725110 DOI: 10.7314/apjcp.2013.14.4.2185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Gallbladder carcinoma, the most frequent malignant neoplasm of the biliary tract system, has always been considered to feature late clinical presentation and diagnosis, limited treatment options and an extremely poor prognosis. In recent years, while the incidence of gallbladder cancer has appeared to be on the increase, the available treatment methods have not greatly improved survival of the affected patients. Thus, exploring new therapeutic targets for this devastating disease is an urgent matter at present. Epidemical studies have demonstrated that the incidence of gallbladder carcinoma exhibits a distinct gender bias, affecting females two to three times more than males, pointing to crucial roles of estrogen. It is well known that estrogen acts on target tissues by binding to estrogen receptors (ERs), which are mainly divided into three subtypes, ERα, ERβ and ERγ. ERα and ERβ appear to have overlapping but also unique even opposite biological effects. As important pathogenic mediators, ERs have been considered to relate to several kinds of tumors. In gallbladder carcinoma tissue, ERs have been shown to be positively expressed, and ERs expression levels are associated with differentiation and prognosis of this cancer. Nevertheless, the exact mechanisms of estrogen inducing growth of gallbladder carcinoma remain poorly understood. On the base of the current investigations, we deduce that estrogen participates in promotion of gallbladder carcinoma by influencing the formation of gallstones, stimulating angiogenesis, and promoting abnormal proliferation. Since ERs mediate the carcinogenic actions of estrogen in gallbladder, and therapy targeting ERs may provide new directions for gallbladder carcinoma. Therefore, it should be stressed that ERs are potential therapeutic targets for gallbladder carcinoma.
Collapse
Affiliation(s)
- Ling-Qiang Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|