1
|
Aberdeen GW, Babischkin JS, Pepe GJ, Albrecht ED. Estrogen stimulates fetal vascular endothelial growth factor expression and microvascularization. J Endocrinol 2024; 262:e230364. [PMID: 38738915 PMCID: PMC11227038 DOI: 10.1530/joe-23-0364] [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: 12/04/2023] [Accepted: 05/12/2024] [Indexed: 05/14/2024]
Abstract
We recently showed that the ratio of capillaries to myofibers in skeletal muscle, which accounts for 80% of insulin-directed glucose uptake and metabolism, was reduced in baboon fetuses in which estrogen was suppressed by maternal letrozole administration. Since vascular endothelial growth factor (VEGF) promotes angiogenesis, the present study determined the impact of estrogen deprivation on fetal skeletal muscle VEGF expression, capillary development, and long-term vascular and metabolic function in 4- to 8-year-old adult offspring. Maternal baboons were untreated or treated with letrozole or letrozole plus estradiol on days 100-164 of gestation (term = 184 days). Skeletal muscle VEGF protein expression was suppressed by 45% (P < 0.05) and correlated (P = 0.01) with a 47% reduction (P < 0.05) in the number of capillaries per myofiber area in fetuses of baboons in which serum estradiol levels were suppressed 95% (P < 0.01) by letrozole administration. The reduction in fetal skeletal muscle microvascularization was associated with a 52% decline (P = 0.02) in acetylcholine-induced brachial artery dilation and a 23% increase (P = 0.01) in mean arterial blood pressure in adult progeny of letrozole-treated baboons, which was restored to normal by letrozole plus estradiol. The present study indicates that estrogen upregulates skeletal muscle VEGF expression and systemic microvessel development within the fetus as an essential programming event critical for ontogenesis of systemic vascular function and insulin sensitivity/glucose homeostasis after birth in primate offspring.
Collapse
Affiliation(s)
- Graham W Aberdeen
- Departments of Obstetrics, Gynecology, Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jeffery S Babischkin
- Departments of Obstetrics, Gynecology, Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Eugene D Albrecht
- Departments of Obstetrics, Gynecology, Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Bai J, Li Y, Yan G, Zhou J, Salmeron AG, Fategbe OT, Kumar S, Chen X, Chen DB. ICI 182,780 Attenuates Selective Upregulation of Uterine Artery Cystathionine β-Synthase Expression in Rat Pregnancy. Int J Mol Sci 2023; 24:14384. [PMID: 37762687 PMCID: PMC10532247 DOI: 10.3390/ijms241814384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/16/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Endogenous hydrogen sulfide (H2S) produced by cystathionine β-synthase (CBS) and cystathionine-γ lyase (CSE) has emerged as a novel uterine vasodilator contributing to pregnancy-associated increases in uterine blood flow, which safeguard pregnancy health. Uterine artery (UA) H2S production is stimulated via exogenous estrogen replacement and is associated with elevated endogenous estrogens during pregnancy through the selective upregulation of CBS without altering CSE. However, how endogenous estrogens regulate uterine artery CBS expression in pregnancy is unknown. This study was conducted to test a hypothesis that endogenous estrogens selectively stimulate UA CBS expression via specific estrogen receptors (ER). Treatment with E2β (0.01 to 100 nM) stimulated CBS but not CSE mRNA in organ cultures of fresh UA rings from both NP and P (gestational day 20, GD20) rats, with greater responses to all doses of E2β tested in P vs. NP UA. ER antagonist ICI 182,780 (ICI, 1 µM) completely attenuated E2β-stimulated CBS mRNA in both NP and P rat UA. Subcutaneous injection with ICI 182,780 (0.3 mg/rat) of GD19 P rats for 24 h significantly inhibited UA CBS but not mRNA expression, consistent with reduced endothelial and smooth muscle cell CBS (but not CSE) protein. ICI did not alter mesenteric and renal artery CBS and CSE mRNA. In addition, ICI decreased endothelial nitric oxide synthase mRNA in UA but not in mesenteric or renal arteries. Thus, pregnancy-augmented UA CBS/H2S production is mediated by the actions of endogenous estrogens via specific ER in pregnant rats.
Collapse
Affiliation(s)
- Jin Bai
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Yao Li
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Guofeng Yan
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Jing Zhou
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Alejandra Garcia Salmeron
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Olamide Tolulope Fategbe
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Sathish Kumar
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Xuejin Chen
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Dong-Bao Chen
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| |
Collapse
|
3
|
Arcadio F, Seggio M, Zeni L, Bossi AM, Cennamo N. Estradiol Detection for Aquaculture Exploiting Plasmonic Spoon-Shaped Biosensors. BIOSENSORS 2023; 13:bios13040432. [PMID: 37185507 PMCID: PMC10136336 DOI: 10.3390/bios13040432] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023]
Abstract
In this work, a surface plasmon resonance (SPR) biosensor based on a spoon-shaped waveguide combined with an estrogen receptor (ERα) was developed and characterized for the detection and the quantification of estradiol in real water samples. The fabrication process for realizing the SPR platform required a single step consisting of metal deposition on the surface of a polystyrene spoon-shaped waveguide featuring a built-in measuring cell. The biosensor was achieved by functionalizing the bowl sensitive surface with a specific estrogen receptor (ERα) that was able to bind the estradiol. In a first phase, the biosensor tests were performed in a phosphate buffer solution obtaining a limit of detection (LOD) equal to 0.1 pM. Then, in order to evaluate the biosensor's response in different real matrices related to aquaculture, its performances were examined in seawater and freshwater. The experimental results support the possibility of using the ERα-based biosensor for the screening of estradiol in both matrices.
Collapse
Affiliation(s)
- Francesco Arcadio
- Department of Engineering, University of Campania Luigi Vanvitelli, Via Roma 29, 81031 Aversa, Italy
| | - Mimimorena Seggio
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134 Verona, Italy
| | - Luigi Zeni
- Department of Engineering, University of Campania Luigi Vanvitelli, Via Roma 29, 81031 Aversa, Italy
| | - Alessandra Maria Bossi
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134 Verona, Italy
| | - Nunzio Cennamo
- Department of Engineering, University of Campania Luigi Vanvitelli, Via Roma 29, 81031 Aversa, Italy
| |
Collapse
|
4
|
Kaur S, Hickman TM, Lopez-Ramirez A, McDonald H, Lockhart LM, Darwish O, Averitt DL. Estrogen modulation of the pronociceptive effects of serotonin on female rat trigeminal sensory neurons is timing dependent and dosage dependent and requires estrogen receptor alpha. Pain 2022; 163:e899-e916. [PMID: 35121697 PMCID: PMC9288423 DOI: 10.1097/j.pain.0000000000002604] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/28/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT The role of the major estrogen estradiol (E2) on orofacial pain conditions remains controversial with studies reporting both a pronociceptive and antinociceptive role of E2. E2 modulation of peripheral serotonergic activity may be one mechanism underlying the female prevalence of orofacial pain disorders. We recently reported that female rats in proestrus and estrus exhibit greater serotonin (5HT)-evoked orofacial nocifensive behaviors compared with diestrus and male rats. Further coexpression of 5HT 2A receptor mRNA in nociceptive trigeminal sensory neurons that express transient receptor potential vanilloid 1 ion channels contributes to pain sensitization. E2 may exacerbate orofacial pain through 5HT-sensitive trigeminal nociceptors, but whether low or high E2 contributes to orofacial pain and by what mechanism remains unclear. We hypothesized that steady-state exposure to a proestrus level of E2 exacerbates 5HT-evoked orofacial nocifensive behaviors in female rats, explored the transcriptome of E2-treated female rats, and determined which E2 receptor contributes to sensitization of female trigeminal sensory neurons. We report that a diestrus level of E2 is protective against 5HT-evoked orofacial pain behaviors, which increase with increasing E2 concentrations, and that E2 differentially alters several pain genes in the trigeminal ganglia. Furthermore, E2 receptors coexpressed with 5HT 2A and transient receptor potential vanilloid 1 and enhanced capsaicin-evoked signaling in the trigeminal ganglia through estrogen receptor α. Overall, our data indicate that low, but not high, physiological levels of E2 protect against orofacial pain, and we provide evidence that estrogen receptor α receptor activation, but not others, contributes to sensitization of nociceptive signaling in trigeminal sensory neurons.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Department of Biology, Texas Woman’s University, Denton, TX 76204
| | | | | | - Hanna McDonald
- Department of Biology, Texas Woman’s University, Denton, TX 76204
| | | | - Omar Darwish
- Department of Mathematics and Computer Science, Texas Woman’s University, Denton, TX 76204
| | | |
Collapse
|
5
|
Wright EB, Lannigan DA. ERK1/2‐RSK regulation of oestrogen homeostasis. FEBS J 2022; 290:1943-1953. [PMID: 35176205 PMCID: PMC9381647 DOI: 10.1111/febs.16407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/23/2021] [Accepted: 02/15/2022] [Indexed: 11/28/2022]
Abstract
The molecular mechanisms regulating oestrogen homeostasis have been primarily studied in the mammary gland, which is the focus of this review. In the non-pregnant adult, the mammary gland undergoes repeated cycles of proliferation and apoptosis in response to the fluctuating levels of oestrogen that occur during the reproductive stage. Oestrogen actions are mediated through the steroid hormone receptors, oestrogen receptor α and β and through a G-protein coupled receptor. In the mammary gland, ERα is of particular importance and thus will be highlighted. Mechanisms regulating oestrogen-induced responses through ERα are necessary to maintain homeostasis given that the signalling pathways that are activated in response to ERα-mediated transcription can also induce transformation. ERK1/2 and its downstream effector, p90 ribosomal S6 kinase (RSK), control homeostasis in the mammary gland by limiting oestrogen-mediated ERα responsiveness. ERK1/2 drives degradation coupled ERα-mediated transcription, whereas RSK2 acts as a negative regulator of ERK1/2 activity to limit oestrogen responsiveness. Moreover, RSK2 acts as a positive regulator of translation. Thus, RSK2 provides both positive and negative signals to maintain oestrogen responsiveness. In addition to transmitting signals through tyrosine kinase receptors, ERK1/2-RSK engages with hedgehog signalling to maintain oestrogen levels and with the HIPPO pathway to regulate ERα-mediated transcription. Additionally, ERK1/2-RSK controls the progenitor populations within the mammary gland to maintain the ERα-positive population. RSK2 is involved in increased breast cancer risk in individuals taking oral contraceptives and in parity-induced protection against breast cancer. RSK2 and ERα may also co-operate in diseases in tissues outside of the mammary gland.
Collapse
Affiliation(s)
- Eric B. Wright
- Biomedical Engineering Vanderbilt University Nashville TN USA
| | - Deborah A. Lannigan
- Biomedical Engineering Vanderbilt University Nashville TN USA
- Pathology, Microbiology & Immunology Vanderbilt University Medical Center Nashville TN USA
- Cell and Developmental Biology Vanderbilt University Nashville TN USA
| |
Collapse
|
6
|
Acramel A, Jacquot Y. Deciphering of a Putative GPER Recognition Domain in ERα and ERα36. Front Endocrinol (Lausanne) 2022; 13:943343. [PMID: 35846328 PMCID: PMC9279910 DOI: 10.3389/fendo.2022.943343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Alexandre Acramel
- CiTCoM laboratory, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8038, Institut National de la Santé et de la Recherche Médicale (INSERM) U1268, Faculty of Pharmacy of Paris, Université Paris Cité, Paris, France
- Department of Pharmacy, Institut Curie, Paris, France
| | - Yves Jacquot
- CiTCoM laboratory, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8038, Institut National de la Santé et de la Recherche Médicale (INSERM) U1268, Faculty of Pharmacy of Paris, Université Paris Cité, Paris, France
- *Correspondence: Yves Jacquot,
| |
Collapse
|
7
|
Adlanmerini M, Fontaine C, Gourdy P, Arnal JF, Lenfant F. Segregation of nuclear and membrane-initiated actions of estrogen receptor using genetically modified animals and pharmacological tools. Mol Cell Endocrinol 2022; 539:111467. [PMID: 34626731 DOI: 10.1016/j.mce.2021.111467] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022]
Abstract
Estrogen receptor alpha (ERα) and beta (ERβ) are members of the nuclear receptor superfamily, playing widespread functions in reproductive and non-reproductive tissues. Beside the canonical function of ERs as nuclear receptors, in this review, we summarize our current understanding of extra-nuclear, membrane-initiated functions of ERs with a specific focus on ERα. Over the last decade, in vivo evidence has accumulated to demonstrate the physiological relevance of this ERα membrane-initiated-signaling from mouse models to selective pharmacological tools. Finally, we discuss the perspectives and future challenges opened by the integration of extra-nuclear ERα signaling in physiology and pathology of estrogens.
Collapse
Affiliation(s)
- Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France.
| |
Collapse
|
8
|
Fiocchetti M, Bastari G, Cipolletti M, Leone S, Acconcia F, Marino M. The Peculiar Estrogenicity of Diethyl Phthalate: Modulation of Estrogen Receptor α Activities in the Proliferation of Breast Cancer Cells. TOXICS 2021; 9:237. [PMID: 34678933 PMCID: PMC8538674 DOI: 10.3390/toxics9100237] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022]
Abstract
Phthalates comprise a group of synthetic chemicals present in the environment because of their wide use as plasticizers and as additives in products for personal care. Among others, diethyl phthalate (DEP) is largely used in products for infants, children, and adults, in which its exposure has been correlated with an increased risk of breast cancer. The adverse health outcomes deriving from phthalate exposure have been associated with their activity as endocrine disruptors (EDCs) of the steroid and thyroid hormone signaling by affecting developmental and reproductive health, and even carcinogenicity. However, the estrogen disruptor activities of DEP are still controversial, and the mechanism at the root of the estrogenic-disrupting action of DEP remains to be clarified. Here, we evaluated the DEP mechanism of action on the activation status of estrogen receptor α (ERα) by analyzing the receptor's phosphorylation as well as both nuclear and extra-nuclear pathways triggered by the receptor to modulate the proliferation of breast cancer cells. Although DEP does not bind to ERα, our results suggest that this phthalate ester exerts multiple parallel interactions with ERα signaling and emphasize the importance to determine an appropriate battery of in vitro methods that will include specific molecular mechanisms involved in the endocrine disruption.
Collapse
Affiliation(s)
- Marco Fiocchetti
- Department of Science, University Roma Tre, Viale G. Marconi, 446, 00146 Rome, Italy; (G.B.); (M.C.); (S.L.); (F.A.)
| | | | | | | | | | - Maria Marino
- Department of Science, University Roma Tre, Viale G. Marconi, 446, 00146 Rome, Italy; (G.B.); (M.C.); (S.L.); (F.A.)
| |
Collapse
|
9
|
Single-cell immunoblotting resolves estrogen receptor-α isoforms in breast cancer. PLoS One 2021; 16:e0254783. [PMID: 34314438 PMCID: PMC8315538 DOI: 10.1371/journal.pone.0254783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/28/2021] [Indexed: 12/18/2022] Open
Abstract
An array of isoforms of the nuclear estrogen receptor alpha (ER-α) protein contribute to heterogeneous response in breast cancer (BCa); yet, a single-cell analysis tool that distinguishes the full-length ER-α66 protein from the activation function-1 deficient ER-α46 isoform has not been reported. Specific detection of protein isoforms is a gap in single-cell analysis tools, as the de facto standard immunoassay requires isoform-specific antibody probes. Consequently, to scrutinize hormone response heterogeneity among BCa tumor cells, we develop a precision tool to specifically measure ER-α66, ER- α46, and eight ER-signaling proteins with single-cell resolution in the highly hetero-clonal MCF-7 BCa cell line. With a literature-validated pan-ER immunoprobe, we distinguish ER-α66 from ER-α46 in each individual cell. We identify ER-α46 in 5.5% of hormone-sensitive (MCF-7) and 4.2% of hormone-insensitive (MDA-MB-231) BCa cell lines. To examine whether the single-cell immunoblotting can capture cellular responses to hormones, we treat cells with tamoxifen and identify different sub-populations of ER-α46: (i) ER-α46 induces phospho-AKT at Ser473, (ii) S6-ribosomal protein, an upstream ER target, activates both ER-α66 and ER-α46 in MCF-7 cells, and (iii) ER-α46 partitions MDA-MB-231 subpopulations, which are responsive to tamoxifen. Unlike other single-cell immunoassays, multiplexed single-cell immunoblotting reports–in the same cell–tamoxifen effects on ER signaling proteins and on distinct isoforms of the ER-α protein.
Collapse
|
10
|
Debarba LK, Jayarathne HSM, Miller RA, Garratt M, Sadagurski M. 17-a-estradiol has sex-specific effects on neuroinflammation that are partly reversed by gonadectomy. J Gerontol A Biol Sci Med Sci 2021; 77:66-74. [PMID: 34309657 DOI: 10.1093/gerona/glab216] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Indexed: 11/13/2022] Open
Abstract
17-α-estradiol (17aE2) treatment from 4-months of age extends lifespan in male mice and can reduce neuroinflammatory responses in the hypothalamus of 12-month-old males. Although 17aE2 improves longevity in males, female mice are unaffected, suggesting a sexually dimorphic pattern of lifespan regulation. We tested whether the sex-specific effects of 17aE2 on neuroinflammatory responses are affected by gonadal removal and whether hypothalamic changes extend to other brain regions in old age. We show that sex-specific effects of 17aE2 on age-associated gliosis are brain region-specific and are partially dependent on gonadectomy. 17aE2 treatment started at 4 months of age protected 25-month-old males from hypothalamic inflammation. Castration before 17aE2 exposure reduced the effect of 17aE2 on hypothalamic astrogliosis in males. By contrast, sex-specific inhibition of microgliosis generated by 17aE2 was not significantly affected by castration. In the hippocampus, gonadectomy influenced the severity of gliosis and the responsiveness to 17aE2 in a region-dependent manner. The male-specific effects of 17aE2 correlate with increases in hypothalamic ERα expression, specifically in gonadally intact males, consistent with the idea that 17aE2 might act through this receptor. Our results indicate that neuroinflammatory responses to 17aE2 are partially controlled by the presence of sex-specific gonads. Loss of gonadal function and age-associated neuroinflammation could, therefore, influence late-life health and disease onset, leading to sexual dimorphism in both aging and in response to drugs that modify the pace of aging.
Collapse
Affiliation(s)
- Lucas K Debarba
- Department of Biological Sciences, IBio (Integrative Biosciences Center), Wayne State University, Detroit, Michigan, MI
| | - Hashan S M Jayarathne
- Department of Biological Sciences, IBio (Integrative Biosciences Center), Wayne State University, Detroit, Michigan, MI
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI
| | - Michael Garratt
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, NZ
| | - Marianna Sadagurski
- Department of Biological Sciences, IBio (Integrative Biosciences Center), Wayne State University, Detroit, Michigan, MI
| |
Collapse
|
11
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
12
|
Gibson DA, Esnal-Zufiaurre A, Bajo-Santos C, Collins F, Critchley HOD, Saunders PTK. Profiling the expression and function of oestrogen receptor isoform ER46 in human endometrial tissues and uterine natural killer cells. Hum Reprod 2021; 35:641-651. [PMID: 32108901 PMCID: PMC7105323 DOI: 10.1093/humrep/dez306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Does the oestrogen receptor isoform, ER46, contribute to regulation of endometrial function? SUMMARY ANSWER ER46 is expressed in endometrial tissues, is the predominant ER isoform in first trimester decidua and is localised to the cell membrane of uterine natural killer (uNK) cells where activation of ER46 increases cell motility. WHAT IS KNOWN ALREADY Oestrogens acting via their cognate receptors are essential regulators of endometrial function and play key roles in establishment of pregnancy. ER46 is a 46-kDa truncated isoform of full length ERα (ER66, encoded by ESR1) that contains both ligand- and DNA-binding domains. Expression of ER46 in the human endometrium has not been investigated previously. ER46 is located at the cell membrane of peripheral blood leukocytes and mediates rapid responses to oestrogens. uNK cells are a phenotypically distinct (CD56brightCD16-) population of tissue-resident immune cells that regulate vascular remodelling within the endometrium and decidua. We have shown that oestrogens stimulate rapid increases in uNK cell motility. Previous characterisation of uNK cells suggests they are ER66-negative, but expression of ER46 has not been characterised. We hypothesise that uNK cells express ER46 and that rapid responses to oestrogens are mediated via this receptor. STUDY DESIGN, SIZE, DURATION This laboratory-based study used primary human endometrial (n = 24) and decidual tissue biopsies (n = 30) as well as uNK cells which were freshly isolated from first trimester human decidua (n = 18). PARTICIPANTS/MATERIALS, SETTING, METHODS Primary human endometrial and first trimester decidual tissue biopsies were collected using methods approved by the local institutional ethics committee (LREC/05/51104/12 and LREC/10/51402/59). The expression of ERs (ER66, ER46 and ERβ) was assessed by quantitative PCR, western blot and immunohistochemistry. uNK cells were isolated from first-trimester human decidua by magnetic bead sorting. Cell motility of uNK cells was measured by live cell imaging: cells were treated with 17β-oestradiol conjugated to bovine serum albumin (E2-BSA, 10 nM equivalent), the ERβ-selective agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN; 10 nM) or dimethylsulphoxide vehicle control. MAIN RESULTS AND THE ROLE OF CHANCE ER46 was detected in proliferative and secretory phase tissues by western blot and was the predominant ER isoform in first-trimester decidua samples. Immunohistochemistry revealed that ER46 was co-localised with ER66 in cell nuclei during the proliferative phase but detected in both the cytoplasm and cell membrane of stromal cells in the secretory phase and in decidua. Triple immunofluorescence staining of decidua tissues identified expression of ER46 in the cell membrane of CD56-positive uNK cells which were otherwise ER66-negative. Profiling of isolated uNK cells confirmed expression of ER46 by quantitative PCR and western blot and localised ER46 protein to the cell membrane by immunocytochemistry. Functional analysis of isolated uNK cells using live cell imaging demonstrated that activation of ER46 with E2-BSA significantly increased uNK cell motility. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Expression pattern in endometrial tissue was only determined using samples from proliferative and secretory phases. Assessment of first trimester decidua samples was from a range of gestational ages, which may have precluded insights into gestation-specific changes in these tissues. Our results are based on in vitro responses of primary human cells and we cannot be certain that similar mechanisms occur in situ. WIDER IMPLICATIONS OF THE FINDINGS E2 is an essential regulator of reproductive competence. This study provides the first evidence for expression of ER46 in the human endometrium and decidua of early pregnancy. We describe a mechanism for regulating the function of human uNK cells via expression of ER46 and demonstrate that selective targeting with E2-BSA regulates uNK cell motility. These novel findings identify a role for ER46 in the human endometrium and provide unique insight into the importance of membrane-initiated signalling in modulating the impact of E2 on uNK cell function in women. Given the importance of uNK cells to regulating vascular remodelling in early pregnancy and the potential for selective targeting of ER46, this may be an attractive future therapeutic target in the treatment of reproductive disorders. STUDY FUNDING/COMPETING INTEREST(S) These studies were supported by Medical Research Council (MRC) Programme Grants G1100356/1 and MR/N024524/1 to PTKS. H.O.D.C. was supported by MRC grant G1002033. The authors declare no competing interests related to the published work.
Collapse
Affiliation(s)
- Douglas A Gibson
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | | | - Cristina Bajo-Santos
- Department of Cancer Research Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Frances Collins
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
13
|
Niță AR, Knock GA, Heads RJ. Signalling mechanisms in the cardiovascular protective effects of estrogen: With a focus on rapid/membrane signalling. Curr Res Physiol 2021; 4:103-118. [PMID: 34746830 PMCID: PMC8562205 DOI: 10.1016/j.crphys.2021.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/22/2022] Open
Abstract
In modern society, cardiovascular disease remains the biggest single threat to life, being responsible for approximately one third of worldwide deaths. Male prevalence is significantly higher than that of women until after menopause, when the prevalence of CVD increases in females until it eventually exceeds that of men. Because of the coincidence of CVD prevalence increasing after menopause, the role of estrogen in the cardiovascular system has been intensively researched during the past two decades in vitro, in vivo and in observational studies. Most of these studies suggested that endogenous estrogen confers cardiovascular protective and anti-inflammatory effects. However, clinical studies of the cardioprotective effects of hormone replacement therapies (HRT) not only failed to produce proof of protective effects, but also revealed the potential harm estrogen could cause. The "critical window of hormone therapy" hypothesis affirms that the moment of its administration is essential for positive treatment outcomes, pre-menopause (3-5 years before menopause) and immediately post menopause being thought to be the most appropriate time for intervention. Since many of the cardioprotective effects of estrogen signaling are mediated by effects on the vasculature, this review aims to discuss the effects of estrogen on vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) with a focus on the role of estrogen receptors (ERα, ERβ and GPER) in triggering the more recently discovered rapid, or membrane delimited (non-genomic), signaling cascades that are vital for regulating vascular tone, preventing hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Ana-Roberta Niță
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
| | - Greg A. Knock
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Richard J. Heads
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
- Cardiovascular Research Section, King’s BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s College London, UK
| |
Collapse
|
14
|
Mann SN, Hadad N, Nelson Holte M, Rothman AR, Sathiaseelan R, Ali Mondal S, Agbaga MP, Unnikrishnan A, Subramaniam M, Hawse J, Huffman DM, Freeman WM, Stout MB. Health benefits attributed to 17α-estradiol, a lifespan-extending compound, are mediated through estrogen receptor α. eLife 2020; 9:59616. [PMID: 33289482 PMCID: PMC7744101 DOI: 10.7554/elife.59616] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic dysfunction underlies several chronic diseases, many of which are exacerbated by obesity. Dietary interventions can reverse metabolic declines and slow aging, although compliance issues remain paramount. 17α-estradiol treatment improves metabolic parameters and slows aging in male mice. The mechanisms by which 17α-estradiol elicits these benefits remain unresolved. Herein, we show that 17α-estradiol elicits similar genomic binding and transcriptional activation through estrogen receptor α (ERα) to that of 17β-estradiol. In addition, we show that the ablation of ERα completely attenuates the beneficial metabolic effects of 17α-E2 in male mice. Our findings suggest that 17α-E2 may act through the liver and hypothalamus to improve metabolic parameters in male mice. Lastly, we also determined that 17α-E2 improves metabolic parameters in male rats, thereby proving that the beneficial effects of 17α-E2 are not limited to mice. Collectively, these studies suggest ERα may be a drug target for mitigating chronic diseases in male mammals.
Collapse
Affiliation(s)
- Shivani N Mann
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Niran Hadad
- The Jackson Laboratory, Bar Harbor, United States
| | - Molly Nelson Holte
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Alicia R Rothman
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Samim Ali Mondal
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Martin-Paul Agbaga
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Archana Unnikrishnan
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | | | - John Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, United States
| | - Willard M Freeman
- Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, United States.,Oklahoma City Veterans Affairs Medical Center, Oklahoma City, United States
| | - Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| |
Collapse
|
15
|
Thiebaut C, Konan HP, Guerquin MJ, Chesnel A, Livera G, Le Romancer M, Dumond H. The Role of ERα36 in Development and Tumor Malignancy. Int J Mol Sci 2020; 21:E4116. [PMID: 32526980 PMCID: PMC7312586 DOI: 10.3390/ijms21114116] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Estrogen nuclear receptors, represented by the canonical forms ERα66 and ERβ1, are the main mediators of the estrogen-dependent pathophysiology in mammals. However, numerous isoforms have been identified, stimulating unconventional estrogen response pathways leading to complex cellular and tissue responses. The estrogen receptor variant, ERα36, was cloned in 2005 and is mainly described in the literature to be involved in the progression of mammary tumors and in the acquired resistance to anti-estrogen drugs, such as tamoxifen. In this review, we will first specify the place that ERα36 currently occupies within the diversity of nuclear and membrane estrogen receptors. We will then report recent data on the impact of ERα36 expression and/or activity in normal breast and testicular cells, but also in different types of tumors including mammary tumors, highlighting why ERα36 can now be considered as a marker of malignancy. Finally, we will explain how studying the regulation of ERα36 expression could provide new clues to counteract resistance to cancer treatments in hormone-sensitive tumors.
Collapse
Affiliation(s)
- Charlène Thiebaut
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| | - Henri-Philippe Konan
- Université de Lyon, F-69000 Lyon, France; (H.-P.K.); (M.L.R.)
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Marie-Justine Guerquin
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiation, Université de Paris, Université Paris Saclay, CEA, F-92265 Fontenay aux Roses, France; (M.-J.G.); (G.L.)
| | - Amand Chesnel
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| | - Gabriel Livera
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiation, Université de Paris, Université Paris Saclay, CEA, F-92265 Fontenay aux Roses, France; (M.-J.G.); (G.L.)
| | - Muriel Le Romancer
- Université de Lyon, F-69000 Lyon, France; (H.-P.K.); (M.L.R.)
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Hélène Dumond
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| |
Collapse
|
16
|
Gutiérrez A, Sambuco L, Álvarez L, Núñez M, Bergoc R, Zotta E, Martín G, Randi A. Expression of estrogen receptor α variants and c-Fos in rat mammary gland and tumors. J Steroid Biochem Mol Biol 2020; 199:105594. [PMID: 31968225 DOI: 10.1016/j.jsbmb.2020.105594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/28/2019] [Accepted: 01/14/2020] [Indexed: 10/25/2022]
Abstract
Breast cancer is currently the leading cause of cancer death among women worldwide. AP-1 (c-Fos/c-Jun) is associated with proliferation and survival, while cytoplasmic c-Fos activates phospholipid synthesis in cells induced to differentiate or grow. Estrogen receptor α 46 (ERα46) is a splice variant of full-length ERα66 and it is known that it has an inhibitory role in cancer cell growth. We investigated c-Fos localization, its relationship to AP-1, the non genomic pathway of phospho-Tyr537-ERα66, as well as ERα46 and ERα66 isoforms in rat mammary gland development and carcinogenic transformation, and in mammary tumors. Female rats were injected: a) saline solution (Control mammary gland, CMG) or b) N-Nitroso-N-methyl urea (NMU), and samples were taken at 60, 90, 120 and 150 days of life. In addition, we analyzed hormone-dependent (HD) and independent (HI) tumors in ovariectomized rats, and intact tumors (IT) in non-ovariectomized ones. Our results show that, in CMG, nuclear c-Fos and proliferation decreased with age, AP-1 content was low, and nuclear ERα46/ERα66 ratio was higher than 1. In NMU, nuclear c-Fos and proliferation increased with carcinogenic transformation, AP-1 content was high, and nuclear ERα46/ERα66 was below 1. As tumor grade increased, proliferation, nuclear c-Fos and AP-1 expression were negatively associated to nuclear ERα46/ERα66 in IT. In HD, nuclear ERα46/ERα66, nuclear c-Fos expression, AP-1 levels and proliferation were lower than in HI, whose growth is estrogen-independent. Phospho-Tyr537-ERα66 content and ERK1/2 activation were associated with AP-1 levels and cell proliferation. Collectively, our findings support the notion that variant detection and ERα46/ERα66 ratio could shed light on the role of ERα isoforms in mammary gland transformation and the behavior of ERα positive mammary tumors.
Collapse
Affiliation(s)
- Alicia Gutiérrez
- Uiversidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5to Piso, (CP1121), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Físicomatematica, Laboratorio de Radioisótopos, Junín 954, Subsuelo, (CP1113), Buenos Aires, Argentina.
| | - Lorena Sambuco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Físicomatematica, Laboratorio de Radioisótopos, Junín 954, Subsuelo, (CP1113), Buenos Aires, Argentina.
| | - Laura Álvarez
- Uiversidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5to Piso, (CP1121), Buenos Aires, Argentina.
| | - Mariel Núñez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Físicomatematica, Laboratorio de Radioisótopos, Junín 954, Subsuelo, (CP1113), Buenos Aires, Argentina.
| | - Rosa Bergoc
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Físicomatematica, Laboratorio de Radioisótopos, Junín 954, Subsuelo, (CP1113), Buenos Aires, Argentina.
| | - Elsa Zotta
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Ciencias Fisiológicas, Sección Patología, Laboratorio de Fisiopatogenia, Paraguay 2155, 5º Piso, (CP1121) Buenos Aires, Argentina.
| | - Gabriela Martín
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Físicomatematica, Laboratorio de Radioisótopos, Junín 954, Subsuelo, (CP1113), Buenos Aires, Argentina.
| | - Andrea Randi
- Uiversidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5to Piso, (CP1121), Buenos Aires, Argentina.
| |
Collapse
|
17
|
Tian W, Wang ZW, Yuan BM, Bao YG. Calycosin induces apoptosis in osteosarcoma cell line via ERβ‑mediated PI3K/Akt signaling pathways. Mol Med Rep 2020; 21:2349-2356. [PMID: 32236598 PMCID: PMC7185272 DOI: 10.3892/mmr.2020.11039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Previous studies have shown that calycosin, a natural phytoestrogen which is structurally similar to estrogen, inhibits proliferation and induces apoptosis in estrogen-dependent cancer types via the estrogen receptor (ER)β-induced inhibition of PI3K/Akt. Therefore, the aims of the present study were to investigate the effects of calycosin on human osteosarcoma (OS), and to examine the molecular mechanisms associated with ERβ. Human OS MG-63 cells were treated with various concentrations of calycosin, and MTT and flow cytometry assays were used to assess the effects of calycosin on cellular proliferation and apoptosis. In addition, protein expression levels of ERβ, phosphorylated (p)-PI3K, p-Akt, cleaved poly (ADP-ribose) polymerase 1 (PARP) and cleaved caspase-3 were evaluated by western blot analysis. The present results suggested that calycosin inhibited proliferation and induced apoptosis in MG-63 cells. Furthermore, increased ERβ expression was detected in OS MG-63 cells treated with calycosin, and an ERβ inhibitor (PHTPP) reversed calycosin-induced cytotoxicity and apoptosis. Moreover, phosphorylation levels of PI3K and Akt were significantly downregulated after calycosin treatment, whereas PHTPP reversed their phosphorylation. ERβ-mediated PI3K/Akt downstream signaling pathways were found to influence the activity of poly (ADP-ribose) polymerase 1 and caspase-3. Thus, the present results indicated that calycosin inhibited proliferation and induced apoptosis in OS MG-63 cells, and that these effects were mediated by ERβ-dependent inhibition of the PI3K/Akt pathways.
Collapse
Affiliation(s)
- Wei Tian
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Zhi-Wei Wang
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Bao-Ming Yuan
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Yong-Ge Bao
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| |
Collapse
|
18
|
Maglione A, Rolla S, Mercanti SFD, Cutrupi S, Clerico M. The Adaptive Immune System in Multiple Sclerosis: An Estrogen-Mediated Point of View. Cells 2019; 8:E1280. [PMID: 31635066 PMCID: PMC6829884 DOI: 10.3390/cells8101280] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/09/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic central nervous system inflammatory disease that leads to demyelination and neurodegeneration. The third trimester of pregnancy, which is characterized by high levels of estrogens, has been shown to be associated with reduced relapse rates compared with the rates before pregnancy. These effects could be related to the anti-inflammatory properties of estrogens, which orchestrate the reshuffling of the immune system toward immunotolerance to allow for fetal growth. The action of these hormones is mediated by the transcriptional regulation activity of estrogen receptors (ERs). Estrogen levels and ER expression define a specific balance of immune cell types. In this review, we explore the role of estradiol (E2) and ERs in the adaptive immune system, with a focus on estrogen-mediated cellular, molecular, and epigenetic mechanisms related to immune tolerance and neuroprotection in MS. The epigenome dynamics of immune systems are described as key molecular mechanisms that act on the regulation of immune cell identity. This is a completely unexplored field, suggesting a future path for more extensive research on estrogen-induced coregulatory complexes and molecular circuitry as targets for therapeutics in MS.
Collapse
Affiliation(s)
- Alessandro Maglione
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy.
| | - Simona Rolla
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy.
| | | | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy.
| | - Marinella Clerico
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy.
| |
Collapse
|
19
|
Rosenfeld CS, Cooke PS. Endocrine disruption through membrane estrogen receptors and novel pathways leading to rapid toxicological and epigenetic effects. J Steroid Biochem Mol Biol 2019; 187:106-117. [PMID: 30465854 PMCID: PMC6370520 DOI: 10.1016/j.jsbmb.2018.11.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/31/2018] [Accepted: 11/18/2018] [Indexed: 01/08/2023]
Abstract
Estrogen binding to estrogen receptors (ESR) triggers signaling cascades within cells. Historically, a major emphasis has been characterizing estrogen-induced genomic actions resulting from binding to nuclear estrogen receptor 1 (nESR1). However, recent evidence indicates the first receptors estrogens encounter as they enter a cell, membrane ESR1 (mESR1), also play crucial roles. Membrane and nuclear ESR are derived from the same transcripts but the former are directed to the membrane via palmitoylation. Binding and activation of mESR1 leads to rapid fluctuations in cAMP and Ca+2 and stimulation of protein kinase pathways. Endocrine disrupting chemicals (EDC) that mimic 17β-estradiol can signal through mESR1 and elicit non-genomic effects. Most current EDC studies have focused on genomic actions via nESR1. However, increasing number of studies have begun to examine potential EDC effects mediated through mESR1, and some EDC might have higher potency for signaling through mESR1 than nESR1. The notion that such chemicals might also affect mESR1 signaling via palmitoylation and depalmitoylation pathways has also begun to gain currency. Recent development of transgenic mice that lack either mESR1 or nESR1, while retaining functional ESR1 in the other compartment, will allow more precise in vivo approaches to determine EDC effects through nESR1 and/or mESR1. It is increasingly becoming apparent in this quickly evolving field that EDC directly affect mESR and estrogen signaling, but such chemicals can also affect proportion of ESR reaching the membrane. Future EDC studies should be designed to consider the full range of effects through mESR alone and in combination with nESR.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA; Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA; Thompson Center for Autism and Neurobehavioral Disorders, Columbia, MO, 65211, USA.
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
20
|
Balogh A, Karpati E, Schneider AE, Hetey S, Szilagyi A, Juhasz K, Laszlo G, Hupuczi P, Zavodszky P, Papp Z, Matko J, Than NG. Sex hormone-binding globulin provides a novel entry pathway for estradiol and influences subsequent signaling in lymphocytes via membrane receptor. Sci Rep 2019; 9:4. [PMID: 30626909 PMCID: PMC6327036 DOI: 10.1038/s41598-018-36882-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 11/22/2018] [Indexed: 02/07/2023] Open
Abstract
The complex effects of estradiol on non-reproductive tissues/cells, including lymphoid tissues and immunocytes, have increasingly been explored. However, the role of sex hormone binding globulin (SHBG) in the regulation of these genomic and non-genomic actions of estradiol is controversial. Moreover, the expression of SHBG and its internalization by potential receptors, as well as the influence of SHBG on estradiol uptake and signaling in lymphocytes has remained unexplored. Here, we found that human and mouse T cells expressed SHBG intrinsically. In addition, B lymphoid cell lines as well as both primary B and T lymphocytes bound and internalized external SHBG, and the amount of plasma membrane-bound SHBG decreased in B cells of pregnant compared to non-pregnant women. As potential mediators of this process, SHBG receptor candidates expressed by lymphocytes were identified in silico, including estrogen receptor (ER) alpha. Furthermore, cell surface-bound SHBG was detected in close proximity to membrane ERs while highly colocalizing with lipid rafts. The SHBG-membrane ER interaction was found functional since SHBG promoted estradiol uptake by lymphocytes and subsequently influenced Erk1/2 phosphorylation. In conclusion, the SHBG-SHBG receptor-membrane ER complex participates in the rapid estradiol signaling in lymphocytes, and this pathway may be altered in B cells in pregnant women.
Collapse
Affiliation(s)
- Andrea Balogh
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary.,Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Eva Karpati
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary.,Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - Szabolcs Hetey
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andras Szilagyi
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Laboratory of Structural Biophysics, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Kata Juhasz
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gloria Laszlo
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary
| | - Petronella Hupuczi
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Peter Zavodszky
- Laboratory of Structural Biophysics, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Janos Matko
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary.
| | - Nandor Gabor Than
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary. .,Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary. .,First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
21
|
Storman EM, Liu NJ, Wessendorf MW, Gintzler AR. Physical Linkage of Estrogen Receptor α and Aromatase in Rat: Oligocrine and Endocrine Actions of CNS-Produced Estrogens. Endocrinology 2018; 159:2683-2697. [PMID: 29771302 PMCID: PMC6692873 DOI: 10.1210/en.2018-00319] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022]
Abstract
Rapid-signaling membrane estrogen receptors (mERs) and aromatase (Aro) are present throughout the central nervous system (CNS), enabling acute regulation of CNS estrogenic signaling. We previously reported that spinal membrane Aro (mAro) and mERα oligomerize (1). As their organizational relationship would likely influence functions of locally produced estrogens, we quantified the mAro and mERα that are physically associated and nonassociated in two functionally different regions of rat CNS: the spinal cord, which has predominantly neural functionalities, and the hypothalamus, which has both neural and endocrine capabilities. Quantitative immunoprecipitation (IP), coimmunoprecipitation, and Western blot analysis were used to quantify the associated and nonassociated subpopulations of mAro and mERα. Regardless of estrous-cycle stage, virtually all mAro was oligomerized with mERα in the spinal cord, whereas only ∼15% was oligomerized in the hypothalamus. The predominance of nonassociated mAro in the hypothalamus, in combination with findings that many hypothalamic Aro-immunoreactive neurons could be retrogradely labeled with peripherally injected Fluoro-Gold, suggests that a portion of hypothalamic estrogens is secreted, potentially regulating pituitary function. Moreover, circulating estrogens increased hypothalamic Aro activity (quantified by the tritiated water-release assay) in the absence of increased Aro protein, revealing nongenomic regulation of Aro activity in the mammalian CNS. The demonstrated presence of associated and nonassociated mAro and mERα subpopulations in the CNS suggests that their selective targeting could restore impaired estrogen-dependent CNS functionalities while minimizing unwanted effects. The full physiological ramifications of brain-secreted estrogens remain to be explored.
Collapse
Affiliation(s)
- Emiliya M Storman
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Nai-Jiang Liu
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Martin W Wessendorf
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Alan R Gintzler
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York
- Correspondence: Alan R. Gintzler, PhD, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, New York 11203. E-mail:
| |
Collapse
|
22
|
Wang Q, Jiang J, Ying G, Xie XQ, Zhang X, Xu W, Zhang X, Song E, Bu H, Ping YF, Yao XH, Wang B, Xu S, Yan ZX, Tai Y, Hu B, Qi X, Wang YX, He ZC, Wang Y, Wang JM, Cui YH, Chen F, Meng K, Wang Z, Bian XW. Tamoxifen enhances stemness and promotes metastasis of ERα36 + breast cancer by upregulating ALDH1A1 in cancer cells. Cell Res 2018; 28:336-358. [PMID: 29393296 PMCID: PMC5835774 DOI: 10.1038/cr.2018.15] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/09/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023] Open
Abstract
The 66 kDa estrogen receptor alpha (ERα66) is the main molecular target for endocrine therapy such as tamoxifen treatment. However, many patients develop resistance with unclear mechanisms. In a large cohort study of breast cancer patients who underwent surgery followed by tamoxifen treatment, we demonstrate that ERα36, a variant of ERα66, correlates with poor prognosis. Mechanistically, tamoxifen directly binds and activates ERα36 to enhance the stemness and metastasis of breast cancer cells via transcriptional stimulation of aldehyde dehydrogenase 1A1 (ALDH1A1). Consistently, the tamoxifen-induced stemness and metastasis can be attenuated by either ALDH1 inhibitors or a specific ERα36 antibody. Thus, tamoxifen acts as an agonist on ERα36 in breast cancer cells, which accounts for hormone therapy resistance and metastasis of breast cancer. Our study not only reveals ERα36 as a stratifying marker for endocrine therapy but also provides a promising therapeutic avenue for tamoxifen-resistant breast cancer.
Collapse
Affiliation(s)
- Qiang Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Jun Jiang
- Department of Breast Diseases, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Guoguang Ying
- Laboratory of Cancer Cell Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Xiao-Qing Xie
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Wei Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xuemin Zhang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, China National Center of Biomedical Analysis, Beijing 100850, China
| | - Erwei Song
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Xiao-Hong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Bin Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Shilei Xu
- Laboratory of Cancer Cell Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Ze-Xuan Yan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Yanhong Tai
- Department of Pathology, General Hospital of PLA, Beijing 100853, China
- Department of Pathology, No.307 Hospital of PLA, Beijing 100071, China
| | - Baoquan Hu
- Department of Breast Diseases, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiaowei Qi
- Department of Breast Diseases, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yan-Xia Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Yan Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - You-Hong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Feng Chen
- Shenogen Pharma Group, Beijing 100085, China
| | - Kun Meng
- Shenogen Pharma Group, Beijing 100085, China
| | - Zhaoyi Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
- Departments of Medical Microbiology & Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE 68178, USA
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| |
Collapse
|
23
|
Rzemieniec J, Litwa E, Wnuk A, Lason W, Kajta M. Bazedoxifene and raloxifene protect neocortical neurons undergoing hypoxia via targeting ERα and PPAR-γ. Mol Cell Endocrinol 2018; 461:64-78. [PMID: 28859903 DOI: 10.1016/j.mce.2017.08.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/04/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022]
Abstract
Selective estrogen receptor modulators (SERMs) such as bazedoxifene and raloxifene are recognized to mainly act via estrogen receptors (ERs), but there is no study examining the involvement of PPAR-γ in their actions, especially in neurons undergoing hypoxia. Little is also known about age-dependent actions of the SERMs on neuronal tissue challenged with hypoxia. In this study, bazedoxifene and raloxifene protected neocortical cells against hypoxia at early and later developmental stages. Both SERMs evoked caspase-3-independent neuroprotection and increased protein levels of ERα (66 and 46 kDa isoforms) and PPAR-γ. In addition, bazedoxifene enhanced expression of ERα-regulated Cyp19a1 mRNA. Using double siRNA silencing, for the first time we demonstrated a key role of ERα and PPAR-γ in the neuroprotective action of the SERMs in neocortical neurons undergoing hypoxia. This study provides prospects for the development of a new therapeutic strategies against hypoxic brain injury that selectively target ERα and/or PPAR-γ.
Collapse
Affiliation(s)
- J Rzemieniec
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - E Litwa
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - A Wnuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - W Lason
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - M Kajta
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland.
| |
Collapse
|
24
|
Machuki J, Zhang H, Harding S, Sun H. Molecular pathways of oestrogen receptors and β-adrenergic receptors in cardiac cells: Recognition of their similarities, interactions and therapeutic value. Acta Physiol (Oxf) 2018; 222. [PMID: 28994249 PMCID: PMC5813217 DOI: 10.1111/apha.12978] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/07/2017] [Accepted: 10/02/2017] [Indexed: 12/18/2022]
Abstract
Oestrogen receptors (ERs) and β-adrenergic receptors (βARs) play important roles in the cardiovascular system. Moreover, these receptors are expressed in cardiac myocytes and vascular tissues. Numerous experimental observations support the hypothesis that similarities and interactions exist between the signalling pathways of ERs (ERα, ERβ and GPR30) and βARs (β1 AR, β2 AR and β3 AR). The recently discovered oestrogen receptor GPR30 shares structural features with the βARs, and this forms the basis for the interactions and functional overlap. GPR30 possesses protein kinase A (PKA) phosphorylation sites and PDZ binding motifs and interacts with A-kinase anchoring protein 5 (AKAP5), all of which enable its interaction with the βAR pathways. The interactions between ERs and βARs occur downstream of the G-protein-coupled receptor, through the Gαs and Gαi proteins. This review presents an up-to-date description of ERs and βARs and demonstrates functional synergism and interactions among these receptors in cardiac cells. We explore their signalling cascades and the mechanisms that orchestrate their interactions and propose new perspectives on the signalling patterns for the GPR30 based on its structural resemblance to the βARs. In addition, we explore the relevance of these interactions to cell physiology, drugs (especially β-blockers and calcium channel blockers) and cardioprotection. Furthermore, a receptor-independent mechanism for oestrogen and its influence on the expression of βARs and calcium-handling proteins are discussed. Finally, we highlight promising therapeutic avenues that can be derived from the shared pathways, especially the phosphatidylinositol-3-OH kinase (PI3K/Akt) pathway.
Collapse
Affiliation(s)
- J.O. Machuki
- Department of Physiology; Xuzhou Medical University; Xuzhou China
| | - H.Y. Zhang
- Department of Physiology; Xuzhou Medical University; Xuzhou China
| | - S.E. Harding
- National Heart and Lung Institute; Imperial College; London UK
| | - H. Sun
- Department of Physiology; Xuzhou Medical University; Xuzhou China
| |
Collapse
|
25
|
Natural Anti-Estrogen Receptor Alpha Antibodies Able to Induce Estrogenic Responses in Breast Cancer Cells: Hypotheses Concerning Their Mechanisms of Action and Emergence. Int J Mol Sci 2018; 19:ijms19020411. [PMID: 29385743 PMCID: PMC5855633 DOI: 10.3390/ijms19020411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 02/04/2023] Open
Abstract
The detection of human anti-estrogen receptor α antibodies (ERαABs) inducing estrogenic responses in MCF-7 mammary tumor cells suggests their implication in breast cancer emergence and/or evolution. A recent report revealing a correlation between the titer of such antibodies in sera from patients suffering from this disease and the percentage of proliferative cells in samples taken from their tumors supports this concept. Complementary evidence of the ability of ERαABs to interact with an epitope localized within the estradiol-binding core of ERα also argues in its favor. This epitope is indeed inserted in a regulatory platform implicated in ERα-initiated signal transduction pathways and transcriptions. According to some experimental observations, two auto-immune reactions may already be advocated to explain the emergence of ERαABs: one involving probably the idiotypic network to produce antibodies acting as estrogenic secretions and the other based on antibodies able to abrogate the action of a natural ERα inhibitor or to prevent the competitive inhibitory potency of released receptor degradation products able to entrap circulating estrogens and co-activators. All of this information, the aspect of which is mainly fundamental, may open new ways in the current tendency to combine immunological and endocrine approaches for the management of breast cancer.
Collapse
|
26
|
Miragem AA, Homem de Bittencourt PI. Nitric oxide-heat shock protein axis in menopausal hot flushes: neglected metabolic issues of chronic inflammatory diseases associated with deranged heat shock response. Hum Reprod Update 2018; 23:600-628. [PMID: 28903474 DOI: 10.1093/humupd/dmx020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/28/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although some unequivocal underlying mechanisms of menopausal hot flushes have been demonstrated in animal models, the paucity of similar approaches in humans impedes further mechanistic outcomes. Human studies might show some as yet unexpected physiological mechanisms of metabolic adaptation that permeate the phase of decreased oestrogen levels in both symptomatic and asymptomatic women. This is particularly relevant because both the severity and time span of hot flushes are associated with increased risk of chronic inflammatory disease. On the other hand, oestrogen induces the expression of heat shock proteins of the 70 kDa family (HSP70), which are anti-inflammatory and cytoprotective protein chaperones, whose expression is modulated by different types of physiologically stressful situations, including heat stress and exercise. Therefore, lower HSP70 expression secondary to oestrogen deficiency increases cardiovascular risk and predisposes the patient to senescence-associated secretory phenotype (SASP) that culminates in chronic inflammatory diseases, such as obesities, type 2 diabetes, neuromuscular and neurodegenerative diseases. OBJECTIVE AND RATIONALE This review focuses on HSP70 and its accompanying heat shock response (HSR), which is an anti-inflammatory and antisenescent pathway whose intracellular triggering is also oestrogen-dependent via nitric oxide (NO) production. The main goal of the manuscript was to show that the vasomotor symptoms that accompany hot flushes may be a disguised clue for important neuroendocrine alterations linking oestrogen deficiency to the anti-inflammatory HSR. SEARCH METHODS Results from our own group and recent evidence on hypothalamic control of central temperature guided a search on PubMed and Google Scholar websites. OUTCOMES Oestrogen elicits rapid production of the vasodilatory gas NO, a powerful activator of HSP70 expression. Whence, part of the protective effects of oestrogen over cardiovascular and neuroendocrine systems is tied to its capacity of inducing the NO-elicited HSR. The hypothalamic areas involved in thermoregulation (infundibular nucleus in humans and arcuate nucleus in other mammals) and whose neurons are known to have their function altered after long-term oestrogen ablation, particularly kisspeptin-neurokinin B-dynorphin neurons, (KNDy) are the same that drive neuroprotective expression of HSP70 and, in many cases, this response is via NO even in the absence of oestrogen. From thence, it is not illogical that hot flushes might be related to an evolutionary adaptation to re-equip the NO-HSP70 axis during the downfall of circulating oestrogen. WIDER IMPLICATIONS Understanding of HSR could shed light on yet uncovered mechanisms of menopause-associated diseases as well as on possible manipulation of HSR in menopausal women through physiological, pharmacological, nutraceutical and prebiotic interventions. Moreover, decreased HSR indices (that can be clinically determined with ease) in perimenopause could be of prognostic value in predicting the moment and appropriateness of starting a HRT.
Collapse
Affiliation(s)
- Antônio Azambuja Miragem
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil.,Federal Institute of Education, Science and Technology 'Farroupilha', Rua Uruguai 1675, Santa Rosa, RS 98900-000, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil
| |
Collapse
|
27
|
Zhang J, Ye ZW, Chen W, Manevich Y, Mehrotra S, Ball L, Janssen-Heininger Y, Tew KD, Townsend DM. S-Glutathionylation of estrogen receptor α affects dendritic cell function. J Biol Chem 2018; 293:4366-4380. [PMID: 29374060 DOI: 10.1074/jbc.m117.814327] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/18/2018] [Indexed: 12/27/2022] Open
Abstract
Glutathione S-transferase Pi (GSTP) is a thiolase that catalyzes the addition of glutathione (GSH) to receptive cysteines in target proteins, producing an S-glutathionylated residue. Accordingly, previous studies have reported that S-glutathionylation is constitutively decreased in cells from mice lacking GSTP (Gstp1/p2-/-). Here, we found that bone marrow-derived dendritic cells (BMDDCs) from Gstp1/p2-/- mice have proliferation rates that are greater than those in their WT counterparts (Gstp1/p2+/+). Moreover, Gstp1/p2-/- BMDDCs had increased reactive oxygen species (ROS) levels and decreased GSH:glutathione disulfide (GSSG) ratios. Estrogen receptor α (ERα) is linked to myeloproliferation and differentiation, and we observed that its steady-state levels are elevated in Gstp1/p2-/- BMDDCs, indicating a link between GSTP and ERα activities. BMDDCs differentiated by granulocyte-macrophage colony-stimulating factor had elevated ERα levels, which were more pronounced in Gstp1/p2-/- than WT mice. When stimulated with lipopolysaccharide for maturation, Gstp1/p2-/- BMDDCs exhibited augmented endocytosis, maturation rate, cytokine secretion, and T-cell activation; heightened glucose uptake and glycolysis; increased Akt signaling (in the mTOR pathway); and decreased AMPK-mediated phosphorylation of proteins. Of note, GSTP formed a complex with ERα, stimulating ERα S-glutathionylation at cysteines 221, 245, 417, and 447; altering ERα's binding affinity for estradiol; and reducing overall binding potential (receptor density and affinity) 3-fold. Moreover, in Gstp1/p2-/- BMDDCs, ERα S-glutathionylation was constitutively decreased. Taken together, these findings suggest that GSTP-mediated S-glutathionylation of ERα controls BMDDC differentiation and affects metabolic function in dendritic cells.
Collapse
Affiliation(s)
- Jie Zhang
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - Zhi-Wei Ye
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - Wei Chen
- Department of Infectious Disease, the Second Affiliated Hospital of Medical School of the Southeast University, 1-1 Zhongfu Road, Nanjing 210003, China, and
| | - Yefim Manevich
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - Shikhar Mehrotra
- Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Lauren Ball
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - Yvonne Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont 05405
| | - Kenneth D Tew
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics,
| | | |
Collapse
|
28
|
Louie MC, Sevigny MB. Steroid hormone receptors as prognostic markers in breast cancer. Am J Cancer Res 2017; 7:1617-1636. [PMID: 28861319 PMCID: PMC5574935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/05/2017] [Indexed: 06/07/2023] Open
Abstract
Despite the existence of many promising anti-cancer therapies, not all breast cancers are equally treatable, due partly to the fact that focus has been primarily on a few select breast cancer biomarkers- notably ERα, PR and HER2. In cases like triple negative breast cancer (ERα-, PR-, and HER2-), there is a complete lack of available biomarkers for prognosis and therapeutic purposes. The goal of this review is to determine if other steroid receptors, like ERβ and AR, could play a prognostic and/or therapeutic role. Data from various in vitro, in vivo, and clinical breast cancer studies were examined to analyze the presence and function of ERβ, PR, and AR in the presence and absence of ERα. Additionally, we focused on studies that examined how expression of the various steroid receptor isoforms affects breast cancer progression. Our findings suggest that while we have a solid understanding of how these receptors work individually, how they interact and behave in the presence and absence of other receptors requires further research. Furthermore, there is an incomplete understanding of how the various steroid receptor isoforms interact and impact receptor function and breast cancer progression, partly due to the difficulty in detecting all the various isoforms. More large-scale clinical studies must be made to analyze systematically the expression of steroid hormone receptors and their respective isoforms in breast cancer patients in order to determine how these receptors interact with each other and in turn affect cancer progression.
Collapse
|
29
|
Miller MM, McMullen PD, Andersen ME, Clewell RA. Multiple receptors shape the estrogen response pathway and are critical considerations for the future of in vitro-based risk assessment efforts. Crit Rev Toxicol 2017; 47:564-580. [DOI: 10.1080/10408444.2017.1289150] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
30
|
Busayapongchai P, Siri S. Simple assay for screening phytoestrogenic compounds using the oestrogen receptor immobilised magnetite nanoparticles. IET Nanobiotechnol 2017; 11:395-402. [PMID: 28530188 DOI: 10.1049/iet-nbt.2016.0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
With increasing interests of phytoestrogens for their potential applications, a rapid and simple tool for screening these phytochemicals is still required. In this study, a simple assay to detect phytoestrogens was developed based on the competition binding between the tested samples and the fluorescently labelled oestrogen (E2) to the human ligand binding domain of oestrogen receptor (LBD-ER) that was immobilised on the magnetite nanoparticles (MNPs). The 40-kDa LBD-ER peptide was produced in an Escherichia coli system. The synthesised 68.7-nm MNPs were silanised and subsequently covalently linked to the C-terminus of LBD-ER peptide. The LBD-ER immobilised MNPs demonstrated the specific binding for the standard E2 with the equilibrium dissociation constant of 9.56 nM and the binding capacity of 0.08 pmol/1 mg of the MNPs. The LBD-ER immobilised MNPs could evaluate oestrogenic activity of the extracts of Asparagus racemosus and Curcuma comosa, the reported phytoestrogenic plants, but not progesterone (P4) and Raphanus sativus extract, the negative controls. The results of this work clearly demonstrated a potential assay for detecting phytoestrogens of crude plant extracts, which is simple and easily adapted to a high throughput format.
Collapse
Affiliation(s)
| | - Sineenat Siri
- School of Biology, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand.
| |
Collapse
|
31
|
Thiebaut C, Chamard-Jovenin C, Chesnel A, Morel C, Djermoune EH, Boukhobza T, Dumond H. Mammary epithelial cell phenotype disruption in vitro and in vivo through ERalpha36 overexpression. PLoS One 2017; 12:e0173931. [PMID: 28301550 PMCID: PMC5354400 DOI: 10.1371/journal.pone.0173931] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/28/2017] [Indexed: 12/16/2022] Open
Abstract
Estrogen receptor alpha 36 (ERα36) is a variant of the canonical estrogen receptor alpha (ERα66), widely expressed in hormone sensitive cancer cells and whose high expression level correlates with a poor survival prognosis for breast cancer patients. While ERα36 activity have been related to breast cancer progression or acquired resistance to treatment, expression level and location of ERα36 are poorly documented in the normal mammary gland. Therefore, we explored the consequences of a ERα36 overexpression in vitro in MCF-10A normal mammary epithelial cells and in vivo in a unique model of MMTV-ERα36 transgenic mouse strain wherein ERα36 mRNA was specifically expressed in the mammary gland. By a combination of bioinformatics and computational analyses of microarray data, we identified hierarchical gene networks, downstream of ERα36 and modulated by the JAK2/STAT3 signaling pathway. Concomitantly, ERα36 overexpression lowered proliferation rate but enhanced migration potential and resistance to staurosporin-induced apoptosis of the MCF-10A cell line. In vivo, ERα36 expression led to duct epithelium thinning and disruption in adult but not in prepubescent mouse mammary gland. These phenotypes correlated with a loss of E-cadherin expression. Here, we show that an enhanced expression of ERα36 is sufficient, by itself, to disrupt normal breast epithelial phenotype in vivo and in vitro through a dominant-positive effect on nongenomic estrogen signaling pathways. These results also suggest that, in the presence of adult endogenous steroid levels, ERα36 overexpression in vivo contributes to alter mammary gland architecture which may support pre-neoplastic lesion and augment breast cancer risk.
Collapse
Affiliation(s)
- Charlène Thiebaut
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Clémence Chamard-Jovenin
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Amand Chesnel
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Chloé Morel
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - El-Hadi Djermoune
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Taha Boukhobza
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Hélène Dumond
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
- * E-mail:
| |
Collapse
|
32
|
Miller MM, Alyea RA, LeSommer C, Doheny DL, Rowley SM, Childs KM, Balbuena P, Ross SM, Dong J, Sun B, Andersen MA, Clewell RA. Editor's Highlight: Development of an In vitro Assay Measuring Uterine-Specific Estrogenic Responses for Use in Chemical Safety Assessment. Toxicol Sci 2016; 154:162-173. [PMID: 27503385 PMCID: PMC5091368 DOI: 10.1093/toxsci/kfw152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
A toxicity pathway approach was taken to develop an in vitro assay using human uterine epithelial adenocarcinoma (Ishikawa) cells as a replacement for measuring an in vivo uterotrophic response to estrogens. The Ishikawa cell was determined to be fit for the purpose of recapitulating in vivo uterine response by verifying fidelity of the biological pathway components and the dose-response predictions to women of child-bearing age. Expression of the suite of estrogen receptors that control uterine proliferation (ERα66, ERα46, ERα36, ERβ, G-protein coupled estrogen receptor (GPER)) were confirmed across passages and treatment conditions. Phenotypic responses to ethinyl estradiol (EE) from transcriptional activation of ER-mediated genes, to ALP enzyme induction and cellular proliferation occurred at concentrations consistent with estrogenic activity in adult women (low picomolar). To confirm utility of this model to predict concentration-response for uterine proliferation with xenobiotics, we tested the concentration-response for compounds with known uterine estrogenic activity in humans and compared the results to assays from the ToxCast and Tox21 suite of estrogen assays. The Ishikawa proliferation assay was consistent with in vivo responses and was a more sensitive measure of uterine response. Because this assay was constructed by first mapping the key molecular events for cellular response, and then ensuring that the assay incorporated these events, the resulting cellular assay should be a reliable tool for identifying estrogenic compounds and may provide improved quantitation of chemical concentration response for in vitro-based safety assessments.
Collapse
Affiliation(s)
- Michelle M Miller
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
- ScitoVation, Research Triangle Park, North Carolina
| | - Rebecca A Alyea
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Caroline LeSommer
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Daniel L Doheny
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
- ScitoVation, Research Triangle Park, North Carolina
| | - Sean M Rowley
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
- ScitoVation, Research Triangle Park, North Carolina
| | - Kristin M Childs
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Pergentino Balbuena
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
- ScitoVation, Research Triangle Park, North Carolina
| | - Susan M Ross
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
- ScitoVation, Research Triangle Park, North Carolina
| | - Jian Dong
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Bin Sun
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Melvin A Andersen
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
- ScitoVation, Research Triangle Park, North Carolina
| | - Rebecca A Clewell
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina;
- ScitoVation, Research Triangle Park, North Carolina
- *The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| |
Collapse
|
33
|
Anderson AM, Ragan MA. Palmitoylation: a protein S-acylation with implications for breast cancer. NPJ Breast Cancer 2016; 2:16028. [PMID: 28721385 PMCID: PMC5515344 DOI: 10.1038/npjbcancer.2016.28] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 12/19/2022] Open
Abstract
Protein S-acylation is a reversible post-translational lipid modification that involves linkage of a fatty acid chain predominantly to a cysteine amino acid via a thioester bond. The fatty acid molecule is primarily palmitate, thus the term 'palmitoylation' is more commonly used. Palmitoylation has been found to modulate all stages of protein function including maturational processing, trafficking, membrane anchoring, signaling range and efficacy, and degradation. In breast cancer, palmitoylation has been shown to control the function of commonly dysregulated genes including estrogen receptors, the epidermal growth factor (EGF) family of receptors, and cancer stem cell markers. Importantly, palmitoylation is a critical factor controlling the formation of complexes at the plasma membrane involving tetraspanins, integrins, and gene products that are key to cell-cell communication. During metastasis, cancer cells enhance their metastatic capacity by interacting with stroma and immune cells. Although aberrant palmitoylation could contribute to tumor initiation and growth, its potential role in these cell-cell interactions is of particular interest, as it may provide mechanistic insight into metastasis, including cancer cell-driven immune modulation. Compelling evidence for a role for aberrant palmitoylation in breast cancer remains to be established. To this end, in this review we summarize emerging evidence and highlight pertinent knowledge gaps, suggesting directions for future research.
Collapse
Affiliation(s)
- Alison M Anderson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Mark A Ragan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
34
|
Clewell RA, McMullen PD, Adeleye Y, Carmichael PL, Andersen ME. Pathway Based Toxicology and Fit-for-Purpose Assays. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 856:205-230. [DOI: 10.1007/978-3-319-33826-2_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
35
|
Adlanmerini M, Fabre A, Boudou F, Riant É, Fontaine C, Laurell H, Gourdy P, Lenfant F, Arnal JF. Effets membranaires du récepteur alpha des œstrogènes. Med Sci (Paris) 2015; 31:1083-91. [DOI: 10.1051/medsci/20153112011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
36
|
Sołtysik K, Czekaj P. ERα36--Another piece of the estrogen puzzle. Eur J Cell Biol 2015; 94:611-25. [PMID: 26522827 DOI: 10.1016/j.ejcb.2015.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 09/03/2015] [Accepted: 10/09/2015] [Indexed: 12/22/2022] Open
Abstract
Although the nuclear action of estrogen receptors (ER) is a well-known fact, evidence supporting membrane estrogen receptors is steadily accumulating. New ER variants of unrecognized function have been discovered. ERα is a product of the ESR1 gene. It serves not only as a template for the full-length 66kDa protein, but also for smaller isoforms which exist as independent receptors. The recently discovered ERα36 (36kDa), consisting of 310 amino acids of total 595 ERα66 protein residues, is an example of that group. The transcription initiation site is identified in the first intron of the ESR1 gene. C-Terminal 27 amino acids are encoded by previously unknown exon 9. The presence of this unique C-terminal sequence creates an opportunity for the production of selective antibodies. ERα36 has been shown to have a high affinity to the cell membrane and as much as 90% of the protein can be bound with it. Post-translational palmitoylation is suspected to play a crucial role in ERα36 anchoring to the cell membrane. In silico analysis suggests the existence of a potential transmembrane domain in ERα36. ERα36 was found in most cells of animals at various ages, but its exact physiological function remains to be fully elucidated. It seems that cells traditionally considered as being deprived of ER are able to respond to hormonal stimulation via the ERα36 receptor. Moreover, ERα36 displays unique pharmacological properties and its action may be behind antiestrogen resistance. The use of ERα36 in cancer diagnosis gives rise to great expectations.
Collapse
Affiliation(s)
- Kamil Sołtysik
- Students Scientific Society, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland.
| |
Collapse
|
37
|
Chuang SC, Chen CH, Fu YC, Tai IC, Li CJ, Chang LF, Ho ML, Chang JK. Estrogen receptor mediates simvastatin-stimulated osteogenic effects in bone marrow mesenchymal stem cells. Biochem Pharmacol 2015; 98:453-64. [PMID: 26410676 DOI: 10.1016/j.bcp.2015.09.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/22/2015] [Indexed: 11/18/2022]
Abstract
Simvastatin, an HMG-CoA reductase inhibitor, is known to promote osteogenic differentiation. However, the mechanism underlying simvastatin-induced osteogenesis is not well understood. In this study, we hypothesize that the estrogen receptor (ER) mediates simvastatin-induced osteogenic differentiation. ER antagonists and siRNA were used to determine the involvement of the ER in simvastatin-induced osteogenesis in mouse bone marrow mesenchymal stem cells (D1 cells). Osteogenesis was evaluated by mRNA expression, protein level/activity of osteogenic markers, and mineralization. The estrogen response element (ERE) promoter activity and the ER-simvastatin binding affinity were examined. Our results showed that the simvastatin-induced osteogenic effects were decreased by treatment with ERα antagonists and ERα siRNA but not by an antagonist specific for the G protein-coupled estrogen receptor (GPER-1). The simvastatin-induced osteogenic effects were further increased by E2 treatment and were reversed by ERα antagonists or siRNA treatment. Luciferase reporter gene assays demonstrated that simvastatin increase ERα-dependent transcriptional activity that was suppressed by ERα antagonists. Furthermore, the ERα-simvastatin binding assay showed that IC50 value of simvastatin is 7.85 μM and that of E2 is 32.8 nM, indicating that simvastatin is a weak ligand for ERα. These results suggest that simvastatin-stimulated osteogenesis is mediated by ERα but not GPER-1. Moreover, this is the first report to demonstrate that simvastatin acts as an ERα ligand and a co-activator to enhance ERα-dependent transcriptional activity and thus promotes osteogenesis. These results indicate that simvastatin-induced osteogenesis is mediated via an ERα-dependent pathway.
Collapse
Affiliation(s)
- Shu-Chun Chuang
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Hwan Chen
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin-Chin Fu
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Chun Tai
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Ju Li
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Fu Chang
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Je-Ken Chang
- Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
38
|
Mata KM, Li W, Reslan OM, Siddiqui WT, Opsasnick LA, Khalil RA. Adaptive increases in expression and vasodilator activity of estrogen receptor subtypes in a blood vessel-specific pattern during pregnancy. Am J Physiol Heart Circ Physiol 2015; 309:H1679-96. [PMID: 26408543 DOI: 10.1152/ajpheart.00532.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/09/2015] [Indexed: 11/22/2022]
Abstract
Normal pregnancy is associated with adaptive hemodynamic, hormonal, and vascular changes, and estrogen (E2) may promote vasodilation during pregnancy; however, the specific E2 receptor (ER) subtype, post-ER signaling mechanism, and vascular bed involved are unclear. We tested whether pregnancy-associated vascular adaptations involve changes in the expression/distribution/activity of distinct ER subtypes in a blood vessel-specific manner. Blood pressure (BP) and plasma E2 were measured in virgin and pregnant (day 19) rats, and the thoracic aorta, carotid artery, mesenteric artery, and renal artery were isolated for measurements of ERα, ERβ, and G protein-coupled receptor 30 [G protein-coupled ER (GPER)] expression and tissue distribution in parallel with relaxation responses to E2 (all ERs) and the specific ER agonist 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)-tris-phenol (PPT; ERα), diarylpropionitrile (DPN; ERβ), and G1 (GPER). BP was slightly lower and plasma E2 was higher in pregnant versus virgin rats. Western blots revealed increased ERα and ERβ in the aorta and mesenteric artery and GPER in the aorta of pregnant versus virgin rats. Immunohistochemistry revealed that the increases in ERs were mainly in the intima and media. In phenylephrine-precontracted vessels, E2 and PPT caused relaxation that was greater in the aorta and mesenteric artery but similar in the carotid and renal artery of pregnant versus virgin rats. DPN- and G1-induced relaxation was greater in the mesenteric and renal artery than in the aorta and carotid artery, and aortic relaxation to G1 was greater in pregnant versus virgin rats. The nitric oxide synthase inhibitor N(ω)-nitro-l-arginine methyl ester with or without the cyclooxygenase inhibitor indomethacin with or without the EDHF blocker tetraethylammonium or endothelium removal reduced E2, PPT, and G1-induced relaxation in the aorta of pregnant rats, suggesting an endothelium-dependent mechanism, but did not affect E2-, PPT-, DPN-, or G1-induced relaxation in other vessels, suggesting endothelium-independent mechanisms. E2, PPT, DPN, and G1 caused relaxation of Ca(2+) entry-dependent KCl contraction, and the effect of PPT was greater in the mesenteric artery of pregnant versus virgin rats. Thus, during pregnancy, an increase in ERα expression in endothelial and vascular smooth muscle layers of the aorta and mesenteric artery is associated with increased ERα-mediated relaxation via endothelium-derived vasodilators and inhibition of Ca(2+) entry into vascular smooth muscle, supporting a role of aortic and mesenteric arterial ERα in pregnancy-associated vasodilation. GPER may contribute to aortic relaxation while enhanced ERβ expression could mediate other genomic vascular effects during pregnancy.
Collapse
Affiliation(s)
- Karina M Mata
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wei Li
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ossama M Reslan
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Waleed T Siddiqui
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lauren A Opsasnick
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
39
|
Bastos CP, Pereira LM, Ferreira-Vieira TH, Drumond LE, Massensini AR, Moraes MFD, Pereira GS. Object recognition memory deficit and depressive-like behavior caused by chronic ovariectomy can be transitorialy recovered by the acute activation of hippocampal estrogen receptors. Psychoneuroendocrinology 2015; 57:14-25. [PMID: 25867995 DOI: 10.1016/j.psyneuen.2015.03.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/18/2015] [Accepted: 03/21/2015] [Indexed: 01/29/2023]
Abstract
It is well known that estradiol (E2) replacement therapy is effective on restoring memory deficits and mood disorders that may occur during natural menopause or after surgical ovarian removal (ovariectomy, OVX). However, it is still unknown the effectiveness of acute and localized E2 administration on the effects of chronic OVX. Here we tested the hypothesis that the intra-hippocampal E2 infusion, as well as specific agonists of estrogen receptors (ERs) alpha (ERα) and beta (ERβ), are able to mend novel object recognition (NOR) memory deficit and depressive-like behavior caused by 12 weeks of OVX. We found that both ERα and ERβ activation, at earlier stages of consolidation, recovered the NOR memory deficit caused by 12 w of OVX. Conversely, only the ERβ activation was effective in decreasing the depressive-like behavior caused by 12 w of OVX. Furthermore, we investigated the effect of OVX on hippocampal volume and ERs expression. The structural MRI showed no alteration in the hippocampus volume of 12 w OVX animals. Interestingly, ERα expression in the hippocampus decreased after one week of OVX, but increased in 12 w OVX animals. Overall, we may conclude that the chronic estrogen deprivation, induced by 12 weeks of OVX, modulates the hippocampal ERα expression and induces NOR memory deficit and depressive-like behaviors. Nonetheless, it is noteworthy that the acute effects of E2 on NOR memory and depressive-like behavior are still apparent even after 12 weeks of OVX.
Collapse
Affiliation(s)
- Cristiane P Bastos
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Brazil
| | - Luciana M Pereira
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Brazil
| | - Talita H Ferreira-Vieira
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Brazil
| | - Luciana E Drumond
- Centro de Tecnologia e Pesquisa em Magneto-Ressonância, CTPMAG, Universidade Federal de Minas Gerais, Brazil; Universidade Federal de São João Del Rey, Brazil
| | - André R Massensini
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Brazil
| | - Márcio F D Moraes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Brazil; Centro de Tecnologia e Pesquisa em Magneto-Ressonância, CTPMAG, Universidade Federal de Minas Gerais, Brazil
| | - Grace S Pereira
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Brazil.
| |
Collapse
|
40
|
Prossnitz ER, Arterburn JB. International Union of Basic and Clinical Pharmacology. XCVII. G Protein-Coupled Estrogen Receptor and Its Pharmacologic Modulators. Pharmacol Rev 2015; 67:505-40. [PMID: 26023144 PMCID: PMC4485017 DOI: 10.1124/pr.114.009712] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Estrogens are critical mediators of multiple and diverse physiologic effects throughout the body in both sexes, including the reproductive, cardiovascular, endocrine, nervous, and immune systems. As such, alterations in estrogen function play important roles in many diseases and pathophysiological conditions (including cancer), exemplified by the lower prevalence of many diseases in premenopausal women. Estrogens mediate their effects through multiple cellular receptors, including the nuclear receptor family (ERα and ERβ) and the G protein-coupled receptor (GPCR) family (GPR30/G protein-coupled estrogen receptor [GPER]). Although both receptor families can initiate rapid cell signaling and transcriptional regulation, the nuclear receptors are traditionally associated with regulating gene expression, whereas GPCRs are recognized as mediating rapid cellular signaling. Estrogen-activated pathways are not only the target of multiple therapeutic agents (e.g., tamoxifen, fulvestrant, raloxifene, and aromatase inhibitors) but are also affected by a plethora of phyto- and xeno-estrogens (e.g., genistein, coumestrol, bisphenol A, dichlorodiphenyltrichloroethane). Because of the existence of multiple estrogen receptors with overlapping ligand specificities, expression patterns, and signaling pathways, the roles of the individual receptors with respect to the diverse array of endogenous and exogenous ligands have been challenging to ascertain. The identification of GPER-selective ligands however has led to a much greater understanding of the roles of this receptor in normal physiology and disease as well as its interactions with the classic estrogen receptors ERα and ERβ and their signaling pathways. In this review, we describe the history and characterization of GPER over the past 15 years focusing on the pharmacology of steroidal and nonsteroidal compounds that have been employed to unravel the biology of this most recently recognized estrogen receptor.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine (E.R.P.) and University of New Mexico Cancer Center (E.R.P., J.B.A.), The University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico (J.B.A.)
| | - Jeffrey B Arterburn
- Department of Internal Medicine (E.R.P.) and University of New Mexico Cancer Center (E.R.P., J.B.A.), The University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico (J.B.A.)
| |
Collapse
|
41
|
Tamoxifen resistance: From cell culture experiments towards novel biomarkers. Pathol Res Pract 2015; 211:189-97. [DOI: 10.1016/j.prp.2015.01.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 12/21/2022]
|
42
|
Nitrate (NO3−) and nitrite (NO2−) are endocrine disruptors to downregulate expression of tyrosine hydroxylase and motor behavior through conversion to nitric oxide in early development of zebrafish. Biochem Biophys Res Commun 2014; 452:608-13. [DOI: 10.1016/j.bbrc.2014.08.114] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 08/22/2014] [Indexed: 12/25/2022]
|
43
|
Wistedt A, Ridderstråle Y, Wall H, Holm L. Exogenous estradiol improves shell strength in laying hens at the end of the laying period. Acta Vet Scand 2014; 56:34. [PMID: 24884886 PMCID: PMC4067625 DOI: 10.1186/1751-0147-56-34] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 05/17/2014] [Indexed: 11/25/2022] Open
Abstract
Background Cracked shells, due to age related reduction of shell quality, are a costly problem for the industry. Parallel to reduced shell quality the skeleton becomes brittle resulting in bone fractures. Calcium, a main prerequisite for both eggshell and bone, is regulated by estrogen in a complex manner. The effects of estrogen, given in a low continuous dose, were studied regarding factors involved in age related changes in shell quality and bone strength of laying hens. A pellet containing 0.385 mg estradiol 3-benzoate (21-day-release) or placebo was inserted subcutaneously in 20 birds each of Lohmann Selected Leghorn (LSL) and Lohmann Brown (LB) at 70 weeks of age. Eggs were collected before and during the experiment for shell quality measurements. Blood samples for analysis of total calcium were taken three days after the insertion and at sacrifice (72 weeks). Right femur was used for bone strength measurements and tissue samples from duodenum and shell gland were processed for morphology, immunohistochemical localization of estrogen receptors (ERα, ERβ), plasma membrane calcium ATPase (PMCA) and histochemical localization of carbonic anhydrase (CA). Results Estrogen treatment increased shell thickness of both hybrids. In addition, shell weight and shell deformation improved in eggs from the brown hybrids. The more pronounced effect on eggs from the brown hybrid may be due to a change in sensitivity to estrogen, especially in surface epithelial cells of the shell gland, shown as an altered ratio between ERα and ERβ. A regulatory effect of estrogen on CA activity, but not PMCA, was seen in both duodenum and shell gland, and a possible connection to shell quality is discussed. Bone strength was unaffected by treatment, but femur was stronger in LSL birds suggesting that the hybrids differ in calcium allocation between shell and bone at the end of the laying period. Plasma calcium concentrations and egg production were unaffected. Conclusions A low continuous dose of estrogen improves shell strength but not bone strength in laying hens at the end of the laying period.
Collapse
|
44
|
Kim KH, Young BD, Bender JR. Endothelial estrogen receptor isoforms and cardiovascular disease. Mol Cell Endocrinol 2014; 389:65-70. [PMID: 24530925 PMCID: PMC4040324 DOI: 10.1016/j.mce.2014.02.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 02/04/2014] [Indexed: 02/07/2023]
Abstract
Rapid, nongenomic vascular cell and tissue responses to estrogen have been demonstrated for more than a decade. Although the pendulum continues to swing, accumulating evidence, both clinical and pre-clinical, support favorable effects of ovarian steroid hormones in the vascular system. These effects are mediated both by classical steroid hormone receptor-mediated transcriptional modulation, and largely by endothelial plasma membrane-associated estrogen receptor (ER)α, which when engaged triggers a signaling cascade resulting in release of cardioprotective nitric oxide (NO). In addition to full-length ERα (ER66), an N-terminus truncated ERα isoform, ER46, plays a key role in these rapid endothelial responses to 17β-estradiol (E2). We have recently determined that ER46 can be a Type I integral transmembrane molecule. In this review, we discuss ER isoforms, rapid E2-stimulated signaling in the endothelium, the importance of the ER46 transmembrane orientation, and the clinical context of this rapid endothelial signaling.
Collapse
Affiliation(s)
- Kyung Hee Kim
- Division of Cardiovascular Medicine and Departments of Internal Medicine and Immunobiology, Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA
| | - Bryan D Young
- Division of Cardiovascular Medicine and Departments of Internal Medicine and Immunobiology, Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA
| | - Jeffrey R Bender
- Division of Cardiovascular Medicine and Departments of Internal Medicine and Immunobiology, Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA.
| |
Collapse
|
45
|
Calycosin suppresses breast cancer cell growth via ERβ-dependent regulation of IGF-1R, p38 MAPK and PI3K/Akt pathways. PLoS One 2014; 9:e91245. [PMID: 24618835 PMCID: PMC3949755 DOI: 10.1371/journal.pone.0091245] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 02/11/2014] [Indexed: 12/27/2022] Open
Abstract
We previously reported that calycosin, a natural phytoestrogen structurally similar to estrogen, successfully triggered apoptosis of estrogen receptor (ER)-positive breast cancer cell line, MCF-7. To better understand the antitumor activities of calycosin against breast cancer, besides MCF-7 cells, another ER-positive cell line T-47D was analyzed here, with ER-negative cell lines (MDA-231, MDA-435) as control. Notably, calycosin led to inhibited cell proliferation and apoptosis only in ER-positive cells, particularly in MCF-7 cells, whereas no such effect was observed in ER-negative cells. Then we investigated whether regulation of ERβ, a subtype of ER, contributed to calycosin-induced apoptosis in breast cancer cells. The results showed that incubation of calycosin resulted in enhanced expression ERβ in MCF-7 and T-47D cells, rather than MDA-231 and MDA-435 cells. Moreover, with the upregulation of ERβ, successive changes in downstream signaling pathways were found, including inactivation of insulin-like growth factor 1 receptor (IGF-1R), then stimulation of p38 MAPK and suppression of the serine/threonine kinase (Akt), and finally poly(ADP-ribose) polymerase 1 (PARP-1) cleavage. However, the other two members of the mitogen-activated protein kinase (MAPK) family, extracellular signal-regulated kinase (ERK) 1/2 and c-Jun N-terminal kinase (JNK), were not consequently regulated by downregulated IGF-1R, indicating ERK 1/2 and JNK pathways were not necessary to allow proliferation inhibition by calycosin. Taken together, our results indicate that calycosin tends to inhibit growth and induce apoptosis in ER-positive breast cancer cells, which is mediated by ERβ-induced inhibition of IGF-1R, along with the selective regulation of MAPK and phosphatidylinositol 3-kinase (PI3K)/Akt pathways.
Collapse
|
46
|
Abstract
Experimental and population-based evidence has been steadily accumulating that steroid hormones are fundamentally involved in the biology of the lung. Both estrogen and progesterone receptors are present in normal and malignant lung tissue, and the reproductive hormones that bind these receptors have a role in lung development, lung inflammation, and lung cancer. The estrogen receptor-β (ER-β) was discovered in the 1990s as a novel form of ER that is transcribed from a gene distinct from ER-α, the receptor previously isolated from breast tissue. Interestingly, ER-β is the predominate ER expressed in normal and malignant lung tissue, whereas inflammatory cells that infiltrate the lung are known to express both ER-α and ER-β. Although there is evidence from animal models for the preferential effects of ER-β in the lungs of females, human lung tumors from males often contain comparable numbers of ER-β-positive cells and male-derived lung cancer cell lines respond to estrogens. Lung tumors from both males and females also express CYP19 (aromatase), the rate-limiting enzyme in estrogen synthesis that converts testosterone to estrone and β-estradiol. Thus, testosterone acts as a precursor for local estrogen production within lung tumors, independent of reproductive organs. This review discusses the recent literature findings about the biology of the ERs, aromatase, and the progesterone receptor in lung cancer and highlights the ongoing clinical trials and future therapeutic implications of these findings.
Collapse
Affiliation(s)
- Jill M Siegfried
- University of Minnesota, 6-120 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455-0217.
| |
Collapse
|
47
|
Abstract
It is becoming increasingly clear that steroid hormones are involved in the biology of many organs outside the reproductive system. Evidence has been accumulating since the mid 1990s that the lung contains receptors for both estrogen and progesterone and that these hormones have some role in lung development, pulmonary inflammation, and lung cancer. The estrogen receptor β (ERβ) is the major ER expressed in lung tissues, while inflammatory cells capable of infiltrating the lung are reported to express both ERα and ERβ. Although there is evidence in animals of preferential effects of ERβ in the lungs of females, human lung tumors from males also contain ERβ-positive cells and express aromatase, the enzyme that converts testosterone to estrogens. This review will discuss current literature findings on the role of the ERs and the progesterone receptor (PR), as well CYP19 (aromatase), the rate-limiting enzyme in the synthesis of estrogen, in lung cancer.
Collapse
Affiliation(s)
- Jill M Siegfried
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA; Current address: Department of Pharmacology, University of Minnesota, Minneapolis, MN.
| | - Laura P Stabile
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|