1
|
Pietrzak-Wawrzyńska BA, Wnuk A, Przepiórska-Drońska K, Łach A, Kajta M. Non-nuclear Estrogen Receptor Signaling as a Promising Therapeutic Target to Reverse Alzheimer's Disease-related Autophagy Deficits and Upregulate the Membrane ESR1 and ESR2 Which Involves DNA Methylation-dependent Mechanisms. J Mol Biol 2025; 437:168982. [PMID: 39914657 DOI: 10.1016/j.jmb.2025.168982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/13/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
Although Alzheimer's disease (AD) affects millions of individuals worldwide, there are currently no effective treatments available. Recent findings have suggested that non-nuclear estrogen receptor (ER) signaling represents promising therapeutic target for central nervous system disorders, offering potential treatments without the significant side effects associated with the activation of nuclear ERs. Because ER signaling deficiency and autophagy impairment have been linked to AD etiology, the present study aimed to selectively target non-nuclear ERs signaling pathways with PaPE-1 and identify autophagy-related mechanisms of neuroprotection in a cellular model of AD. The present study demonstrated that PaPE-1 protected mouse cortical neurons from AD pathology, as evidenced by MAP2-specific labeling. Posttreatment with PaPE-1 reversed the amyloid-β (Aβ)-evoked decrease in autophagic vesicles level, and increased the expression of autophagy-related mRNAs and proteins, accompanied by hypomethylation of the Atg7 gene. Moreover, posttreatment with PaPE-1 increased the levels of membrane fraction receptors ESR1/ERα and ESR2/ERβ, which corresponds to increased Esr1 and Esr2 mRNA expression and DNA hypomethylation of specific genes. In addition to inhibiting DNA methylation of autophagy and ER-related genes, PaPE-1 did not alter global DNA methylation but stimulated HAT activity in Aβ-treated cells. In summary, PaPE-1 promoted neuroprotection against Aβ-induced toxicity that involved stimulation of autophagy, upregulation of membrane ESR1 and ESR2 and decreased DNA methylation of respective genes. The present study proposes a novel therapeutic approach against AD that is based on the selective activation of non-nuclear ER signaling to overcome Aβ-induced autophagy deficits and normalize the epigenetic status of cerebral neurons.
Collapse
Affiliation(s)
- Bernadeta A Pietrzak-Wawrzyńska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12 31-343 Krakow, Poland
| | - Agnieszka Wnuk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12 31-343 Krakow, Poland
| | - Karolina Przepiórska-Drońska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12 31-343 Krakow, Poland
| | - Andrzej Łach
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12 31-343 Krakow, Poland
| | - Małgorzata Kajta
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12 31-343 Krakow, Poland.
| |
Collapse
|
2
|
Oliphant MU, Akshinthala D, Muthuswamy SK. Establishing conditions for the generation and maintenance of estrogen receptor-positive organoid models of breast cancer. RESEARCH SQUARE 2023:rs.3.rs-3341539. [PMID: 37886440 PMCID: PMC10602055 DOI: 10.21203/rs.3.rs-3341539/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Patient-derived organoid models of estrogen receptor-positive (ER+) breast cancer would provide a much-needed tool to understand drug resistance and disease progression better. However, the establishment and long-term maintenance of ER expression, function, and response in vitro remains a significant challenge. Here, we report the development of an ER+ breast tumor organoid medium (BTOM-ER) that conserves ER expression, estrogen responsiveness, and dependence, as well as sensitivity to endocrine therapy of ER+ patient-derived xenograft organoids (PDXO). Our findings demonstrate the utility of subtype-specific culture conditions that better mimic the characteristics of the breast epithelial biology and microenvironment, providing a powerful platform for investigating therapy response and disease progression of ER+ breast cancer.
Collapse
|
3
|
Johnson CS, Mermelstein PG. The interaction of membrane estradiol receptors and metabotropic glutamate receptors in adaptive and maladaptive estradiol-mediated motivated behaviors in females. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:33-91. [PMID: 36868633 DOI: 10.1016/bs.irn.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Estrogen receptors were initially identified as intracellular, ligand-regulated transcription factors that result in genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor α and estrogen receptor β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) can rapidly alter cellular excitability and gene expression, particularly through the phosphorylation of CREB. A principal mechanism of neuronal mER action has been shown to occur through glutamate-independent transactivation of metabotropic glutamate receptors (mGlu), which elicits multiple signaling outcomes. The interaction of mERs with mGlu has been shown to be important in many diverse functions in females, including driving motivated behaviors. Experimental evidence suggests that a large part of estradiol-induced neuroplasticity and motivated behaviors, both adaptive and maladaptive, occurs through estradiol-dependent mER activation of mGlu. Herein we will review signaling through estrogen receptors, both "classical" nuclear receptors and membrane-bound receptors, as well as estradiol signaling through mGlu. We will focus on how the interactions of these receptors and their downstream signaling cascades are involved in driving motivated behaviors in females, discussing a representative adaptive motivated behavior (reproduction) and maladaptive motivated behavior (addiction).
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
4
|
Karst H, den Boon FS, Vervoort N, Adrian M, Kapitein LC, Joëls M. Non-genomic steroid signaling through the mineralocorticoid receptor: Involvement of a membrane-associated receptor? Mol Cell Endocrinol 2022; 541:111501. [PMID: 34740745 DOI: 10.1016/j.mce.2021.111501] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/05/2021] [Accepted: 10/27/2021] [Indexed: 12/25/2022]
Abstract
Corticosteroid receptors in the mammalian brain mediate genomic as well as non-genomic actions. Although receptors mediating genomic actions were already cloned 35 years ago, it remains unclear whether the same molecules are responsible for the non-genomic actions or that the latter involve a separate class of receptors. Here we focus on one type of corticosteroid receptors, i.e. the mineralocorticoid receptor (MR). We summarize some of the known properties and the current insight in the localization of the MR in peripheral cells and neurons, especially in relation to non-genomic signaling. Previous studies from our own and other labs provided evidence that MRs mediating non-genomic actions are identical to the ones involved in genomic signaling, but may be translocated to the plasma cell membrane instead of the nucleus. With fixed cell imaging and live cell imaging techniques we tried to visualize these presumed membrane-associated MRs, using antibodies or overexpression of MR-GFP in COS7 and hippocampal cultured neurons. Despite the physiological evidence for MR location in or close to the cell membrane, we could not convincingly visualize membrane localization of endogenous MRs or GFP-MR molecules. However, we did find punctae of labeled antibodies intracellularly, which might indicate transactivating spots of MR near the membrane. We also found some evidence for trafficking of MR via beta-arrestins. In beta-arrestin knockout mice, we didn't observe metaplasticity in the basolateral amygdala anymore, indicating that internalization of MRs could play a role during corticosterone activation. Furthermore, we speculate that membrane-associated MRs could act indirectly via activating other membrane located structures like e.g. GPER and/or receptor tyrosine kinases.
Collapse
Affiliation(s)
- Henk Karst
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands.
| | - Femke S den Boon
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Niek Vervoort
- University Utrecht, Faculty of Science, Division of Cell Biology, Utrecht, the Netherlands
| | - Max Adrian
- University Utrecht, Faculty of Science, Division of Cell Biology, Utrecht, the Netherlands
| | - Lukas C Kapitein
- University Utrecht, Faculty of Science, Division of Cell Biology, Utrecht, the Netherlands
| | - Marian Joëls
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands; University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
5
|
Clark S, Pollard K, Rainville J, Vasudevan N. Immunoblot Detection of the Phosphorylation of the Estrogen Receptor α as an Outcome of GPR30 /GPER1 Activation. Methods Mol Biol 2022; 2418:25-39. [PMID: 35119657 DOI: 10.1007/978-1-0716-1920-9_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Phosphorylation of the serine residues in estrogen receptor (ER) α is important in transcriptional activation. Hence, methods to detect such posttranslational modification events are valuable. We describe, in detail, the analysis of the phosphorylated ERα by electrophoretic separation of proteins and subsequent immunoblotting techniques. In particular, phosphorylation of the ERα is one possible outcome of activation of the putative membrane estrogen receptor (mER), GPR30 or GPER1. Hence, phosphorylation represents a crosstalk event between GPR30 and ERα and may be important in estrogen-regulated physiology.
Collapse
Affiliation(s)
- Sara Clark
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
| | - Kevin Pollard
- Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Jennifer Rainville
- Department of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | | |
Collapse
|
6
|
Johnson CS, Micevych PE, Mermelstein PG. Membrane estrogen signaling in female reproduction and motivation. Front Endocrinol (Lausanne) 2022; 13:1009379. [PMID: 36246891 PMCID: PMC9557733 DOI: 10.3389/fendo.2022.1009379] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/13/2022] [Indexed: 01/13/2023] Open
Abstract
Estrogen receptors were initially identified in the uterus, and later throughout the brain and body as intracellular, ligand-regulated transcription factors that affect genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor-α and estrogen receptor-β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) not only rapidly effect cellular excitability, but can and do ultimately affect gene expression, as seen through the phosphorylation of CREB. A principal mechanism of neuronal mER action is through glutamate-independent transactivation of metabotropic glutamate receptors (mGluRs), which elicits multiple signaling outcomes. The interaction of mERs with mGluRs has been shown to be important in many diverse functions in females, including, but not limited to, reproduction and motivation. Here we review membrane-initiated estrogen receptor signaling in females, with a focus on the interactions between these mERs and mGluRs.
Collapse
Affiliation(s)
- Caroline S. Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Caroline S. Johnson,
| | - Paul E Micevych
- Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Paul G. Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
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
|
Acconcia F, Fiocchetti M, Busonero C, Fernandez VS, Montalesi E, Cipolletti M, Pallottini V, Marino M. The extra-nuclear interactome of the estrogen receptors: implications for physiological functions. Mol Cell Endocrinol 2021; 538:111452. [PMID: 34500041 DOI: 10.1016/j.mce.2021.111452] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/19/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023]
Abstract
Over the last decades, a great body of evidence has defined a novel view of the cellular mechanism of action of the steroid hormone 17β-estradiol (E2) through its estrogen receptors (i.e., ERα and ERβ). It is now clear that the E2-activated ERs work both as transcription factors and extra-nuclear plasma membrane-localized receptors. The activation of a plethora of signal transduction cascades follows the E2-dependent engagement of plasma membrane-localized ERs and is required for the coordination of gene expression, which ultimately controls the occurrence of the pleiotropic effects of E2. The definition of the molecular mechanisms by which the ERs locate at the cell surface (i.e., palmitoylation and protein association) determined the quest for understanding the specificity of the extra-nuclear E2 signaling. The use of mice models lacking the plasma membrane ERα localization unveiled that the extra-nuclear E2 signaling is operational in vivo but tissue-specific. However, the underlying molecular details for such ERs signaling diversity in the perspective of the E2 physiological functions in the different cellular contexts are still not understood. Therefore, to gain insights into the tissue specificity of the extra-nuclear E2 signaling to physiological functions, here we reviewed the known ERs extra-nuclear interactors and tried to extrapolate from available databases the ERα and ERβ extra-nuclear interactomes. Based on literature data, it is possible to conclude that by specifically binding to extra-nuclear localized proteins in different sub-cellular compartments, the ERs fine-tune their molecular activities. Moreover, we report that the context-dependent diversity of the ERs-mediated extra-nuclear E2 actions can be ascribed to the great flexibility of the physical structures of ERs and the spatial-temporal organization of the logistics of the cells (i.e., the endocytic compartments). Finally, we provide lists of proteins belonging to the potential ERα and ERβ extra-nuclear interactomes and propose that the systematic experimental definition of the ERs extra-nuclear interactomes in different tissues represents the next step for the research in the ERs field. Such characterization will be fundamental for the identification of novel druggable targets for the innovative treatment of ERs-related diseases.
Collapse
Affiliation(s)
- Filippo Acconcia
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy.
| | - Marco Fiocchetti
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Claudia Busonero
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Virginia Solar Fernandez
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Emiliano Montalesi
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Manuela Cipolletti
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Valentina Pallottini
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Maria Marino
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy.
| |
Collapse
|
9
|
Smedlund KB, Hill JW. The role of non-neuronal cells in hypogonadotropic hypogonadism. Mol Cell Endocrinol 2020; 518:110996. [PMID: 32860862 DOI: 10.1016/j.mce.2020.110996] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/01/2020] [Accepted: 08/16/2020] [Indexed: 12/18/2022]
Abstract
The hypothalamic-pituitary-gonadal axis is controlled by gonadotropin-releasing hormone (GnRH) released by the hypothalamus. Disruption of this system leads to impaired reproductive maturation and function, a condition known as hypogonadotropic hypogonadism (HH). Most studies to date have focused on genetic causes of HH that impact neuronal development and function. However, variants may also impact the functioning of non-neuronal cells known as glia. Glial cells make up 50% of brain cells of humans, primates, and rodents. They include radial glial cells, microglia, astrocytes, tanycytes, oligodendrocytes, and oligodendrocyte precursor cells. Many of these cells influence the hypothalamic neuroendocrine system controlling fertility. Indeed, glia regulate GnRH neuronal activity and secretion, acting both at their cell bodies and their nerve endings. Recent work has also made clear that these interactions are an essential aspect of how the HPG axis integrates endocrine, metabolic, and environmental signals to control fertility. Recognition of the clinical importance of interactions between glia and the GnRH network may pave the way for the development of new treatment strategies for dysfunctions of puberty and adult fertility.
Collapse
Affiliation(s)
- Kathryn B Smedlund
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
10
|
Stewart MK, Mattiske DM, Pask AJ. Oestrogen regulates SOX9 bioavailability by rapidly activating ERK1/2 and stabilising microtubules in a human testis-derived cell line. Exp Cell Res 2020; 398:112405. [PMID: 33271127 DOI: 10.1016/j.yexcr.2020.112405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 01/31/2023]
Abstract
Nuclear SOX9 is essential for Sertoli cell differentiation and the development of a testis. Exposure of Sertoli cells to exogenous oestrogen causes cytoplasmic retention of SOX9, inhibiting testis development and promoting ovarian development. The cytoplasmic localisation of SOX9 requires a stabilised microtubule network and a key MAPK complex, ERK1/2, is responsive to oestrogen and known to affect the microtubule network. We hypothesised that oestrogen could stabilise microtubules through the activation of ERK1/2 to promote the cytoplasmic retention of SOX9. Treatment of human testis-derived NT2/D1 cells for 30 min with oestrogen rapidly activated ERK1/2, stabilised the microtubule network and increased cytoplasmic localisation of SOX9. The effects of oestrogen on SOX9 and tubulin were blocked by the ERK1/2 inhibitor U0126, demonstrating that ERK1/2 mediates the stabilisation of microtubules and cytoplasmic retention of SOX9 by oestrogen. Together, these data revealed a previously unknown mechanism for oestrogen in impacting MAPK signalling to block SOX9 bioavailability and the differentiation of Sertoli cells.
Collapse
Affiliation(s)
- Melanie K Stewart
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Deidre M Mattiske
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Vahidinia Z, Karimian M, Joghataei MT. Neurosteroids and their receptors in ischemic stroke: From molecular mechanisms to therapeutic opportunities. Pharmacol Res 2020; 160:105163. [DOI: 10.1016/j.phrs.2020.105163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 01/09/2023]
|
12
|
Cornil CA, Ball GF, Balthazart J. Sexually differentiated and neuroanatomically specific co-expression of aromatase neurons and GAD67 in the male and female quail brain. Eur J Neurosci 2020; 52:2963-2981. [PMID: 32349174 DOI: 10.1111/ejn.14765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/23/2020] [Indexed: 11/30/2022]
Abstract
Testosterone aromatization into estrogens in the preoptic area (POA) is critical for the activation of male sexual behavior in many vertebrates. Yet, the cellular mechanisms mediating actions of neuroestrogens on sexual behavior remain largely unknown. We investigated in male and female Japanese quail by dual-label fluorescent in situ hybridization (FISH) whether aromatase-positive (ARO) neurons express glutamic acid decarboxylase 67 (GAD67), the rate-limiting enzyme in GABA biosynthesis. ARO cells and ARO cells double labeled with GAD67 (ARO-GAD67) were counted at standardized locations in the medial preoptic nucleus (POM) and the medial bed nucleus of the stria terminalis (BST) to produce three-dimensional distribution maps. Overall, males had more ARO cells than females in POM and BST. The number of double-labeled ARO-GAD67 cells was also higher in males than in females and greatly varied as a function of the specific position in these nuclei. Significant sex differences were however present only in the most caudal part of POM. Although both ARO and GAD67 were expressed in the VMN, no colocalization between these markers was detected. Together, these data show that a high proportion of estrogen-synthesizing neurons in POM and BST are inhibitory and the colocalization of GAD67 with ARO exhibits a high degree of anatomical specificity as well as localized sex differences. The fact that many preoptic ARO neurons project to the periaqueductal gray in male quail suggests possible mechanisms through which locally produced estrogens could activate male sexual behavior.
Collapse
Affiliation(s)
| | - Gregory F Ball
- Department of Psychology, University of Maryland, College Park, MD, USA
| | | |
Collapse
|
13
|
Haumann I, Sturm MA, Anstötz M, Rune GM. GPER1 Signaling Initiates Migration of Female V-SVZ-Derived Cells. iScience 2020; 23:101077. [PMID: 32361597 PMCID: PMC7200306 DOI: 10.1016/j.isci.2020.101077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/01/2019] [Accepted: 04/14/2020] [Indexed: 12/30/2022] Open
Abstract
In the rodent ventricular-subventricular zone (V-SVZ) neurons are generated throughout life. They migrate along the rostral migratory stream (RMS) into the olfactory bulb before their final differentiation into interneurons and integration into local circuits. Estrogen receptors (ERs) are steroid hormone receptors with important functions in neurogenesis and synaptic plasticity. In this study, we show that the ER GPER1 is expressed in subsets of cells within the V-SVZ of female animals and provide evidence for a potential local estrogen source from aromatase-positive astrocytes surrounding the RMS. Blocking of GPER1 in Matrigel cultures of female animals significantly impairs migration of V-SVZ-derived cells. This outgrowth is accompanied by regulation of phosphorylation of the actin-binding protein cofilin by GPER1 signaling including an involvement of the p21-Ras pathway. Our results point to a prominent role of GPER1 in the initiation of neuronal migration from the V-SVZ to the olfactory bulb. GPER1 is expressed within all cell types of the stem cell lineage in the V-SVZ Blocking of GPER1 leads to a decrease in migration of V-SVZ-derived neuroblasts GPER1 signaling in V-SVZ Matrigel cultures involves Ras-induced p21 Blocking of GPER1 signaling leads to an increase in the ratio of p-cofilin/cofilin
Collapse
Affiliation(s)
- Iris Haumann
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Muriel Anne Sturm
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Max Anstötz
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
14
|
Wong AM, Scott AK, Johnson CS, Mohr MA, Mittelman-Smith M, Micevych PE. ERαΔ4, an ERα splice variant missing exon4, interacts with caveolin-3 and mGluR2/3. J Neuroendocrinol 2019; 31:e12725. [PMID: 31050077 PMCID: PMC6591055 DOI: 10.1111/jne.12725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/11/2019] [Accepted: 04/26/2019] [Indexed: 12/11/2022]
Abstract
The two isoforms of the nuclear estrogen receptor, ERα and ERβ are widely expressed in the central nervous system. Although they were first described as nuclear receptors, both isoforms have also been found at the cell membrane where they mediate cell signaling. Surface biotinylation studies using neuronal and glial primary cultures label an alternatively spliced form of ERα. The 52 kDa protein, ERαΔ4, is missing exon 4 and is highly expressed in membrane fractions derived from cultured cells. In vivo, both full-length (66 kDa) ERα and ERαΔ4 are present in membrane fractions. In response to estradiol, full-length ERα and ERαΔ4 are initially trafficked to the membrane, and then internalized in parallel. Previous studies determined that only the full-length ERα associates with metabotropic glutamate receptor-1a (mGluR1a), initiating cellular signaling. The role of ERαΔ4, remained to be elucidated. Here, we report ERαΔ4 trafficking, association with mGluR2/3, and downstream signaling in female rat arcuate nucleus (ARH). Caveolin (CAV) proteins are needed for ER transport to the cell membrane, and using co-immunoprecipitation CAV-3 was shown to associate with ERαΔ4. CAV-3 was necessary for ERαΔ4 trafficking to the membrane: in the ARH, microinjection of CAV-3 siRNA reduced CAV-3 and ERαΔ4a in membrane fractions by 50%, and 60%, respectively. Moreover, co-immunoprecipitation revealed that ERαΔ4 associated with inhibitory mGluRs, mGluR2/3. Estrogen benzoate (EB) treatment (5 μg; s.c.; every 4 days; three cycles) reduced levels of cAMP, an effect attenuated by antagonizing mGluR2/3. Following EB treatment, membrane levels of ERαΔ4 and mGluR2/3 were reduced implying ligand-induced internalization. These results implicate ERαΔ4 in an estradiol-induced inhibitory cell signaling in the ARH.
Collapse
Affiliation(s)
- Angela M Wong
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Alexandra K Scott
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Caroline S Johnson
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Margaret A Mohr
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Melinda Mittelman-Smith
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Paul E Micevych
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| |
Collapse
|
15
|
Tominna R, Chokr S, Feri M, Chuon T, Sinchak K. Plasma membrane G protein-coupled estrogen receptor 1 (GPER) mediates rapid estradiol facilitation of sexual receptivity through the orphanin-FQ-ORL-1 system in estradiol primed female rats. Horm Behav 2019; 112:89-99. [PMID: 30981690 DOI: 10.1016/j.yhbeh.2019.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/26/2019] [Accepted: 04/08/2019] [Indexed: 11/29/2022]
Abstract
In estradiol-primed nonreceptive ovariectomized rats, activation of G protein-coupled estrogen receptor 1 (GPER) in the arcuate nucleus of the hypothalamus (ARH) rapidly facilitates sexual receptivity (lordosis). Estradiol priming activates ARH β-endorphin (β-END) neurons that then activate medial preoptic (MPN) μ-opioid receptors (MOP) to inhibit lordosis. ARH infusion of non-esterified 17β-estradiol (E2) 47.5 h after 17β-estradiol benzoate (2 μg EB) priming deactivates MPN MOP and rapidly facilitates lordosis within 30 min via activation of GPER. Since it was unclear where GPERs were located in the neuron, we tested the hypothesis that GPER signaling is initiated at the plasma membrane. Membrane impermeable estradiol (17β-estradiol conjugated to biotin; E-Biotin) infused into the ARH of EB primed rats facilitated lordosis within 30 min, and MPN MOP was deactivated. These actions were blocked by pretreating with GPER antagonist, G-15. Further, we used cell fractionation and western blot techniques to demonstrate that GPER is expressed both in plasma membrane and cytosolic ARH fractions. In previous studies, the orphanin FQ/nociceptin-opioid receptor-like receptor-1 (OFQ/N-ORL-1) system mediated estradiol-only facilitation of lordosis. Therefore, we tested whether the OFQ/N-ORL-1 system mediates E-Biotin-GPER facilitation of lordosis. Pretreatment of UFP-101, an ORL-1 selective antagonist, blocked the facilitation of lordosis and deactivation of MPN MOP by ARH infusion of E-Biotin. Double-label immunohistochemistry revealed that GPER is expressed within approximately 70% of OFQ/N neurons. These data indicate that membrane GPER mediates the E2/E-Biotin facilitation of lordosis by inducing OFQ/N neurotransmission, which inhibits β-END neurotransmission to reduce MPN MOP activation.
Collapse
Affiliation(s)
- Reema Tominna
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA, United States of America
| | - Sima Chokr
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA, United States of America
| | - Micah Feri
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA, United States of America
| | - Timbora Chuon
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA, United States of America
| | - Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA, United States of America.
| |
Collapse
|
16
|
Micevych PE, Sinchak K. Extranuclear signaling by ovarian steroids in the regulation of sexual receptivity. Horm Behav 2018; 104:4-14. [PMID: 29753716 PMCID: PMC6240501 DOI: 10.1016/j.yhbeh.2018.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Paul E Micevych
- Dept of Neurobiology, David Geffen School of Medicine at UCLA, Laboratory of Neuroendocrinology of the UCLA Brain Research Institute, United States
| | - Kevin Sinchak
- Dept of Biological Sciences, California State University, Long Beach, United States.
| |
Collapse
|
17
|
Stincic TL, Rønnekleiv OK, Kelly MJ. Diverse actions of estradiol on anorexigenic and orexigenic hypothalamic arcuate neurons. Horm Behav 2018; 104:146-155. [PMID: 29626486 PMCID: PMC6196116 DOI: 10.1016/j.yhbeh.2018.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 12/13/2022]
Abstract
Contribution to Special Issue on Fast effects of steroids. There is now compelling evidence for membrane-associated estrogen receptors in hypothalamic neurons that are critical for the hypothalamic control of homeostatic functions. It has been known for some time that estradiol (E2) can rapidly alter hypothalamic neuronal activity within seconds, indicating that some cellular effects can occur via membrane initiated events. However, our understanding of how E2 signals via membrane-associated receptors and how these signals impact physiological functions is only just emerging. Thus, E2 can affect second messenger systems including calcium mobilization and a plethora of kinases to alter cell excitability and even gene transcription in hypothalamic neurons. One population of hypothalamic neurons, the anorexigenic proopiomelanocortin (POMC) neurons, has long been considered to be a target of E2's actions based on gene (Pomc) expression studies. However, we now know that E2 can rapidly alter POMC neuronal activity within seconds and activate several intracellular signaling cascades that ultimately affect gene expression, actions which are critical for maintaining sensitivity to insulin in metabolically stressed states. E2 also affects the orexigenic Neuropeptide Y/Agouti-related Peptide (NPY/AgRP) neurons in similarly rapid but antagonistic manner. Therefore, this review will summarize our current state of knowledge of how E2 signals via rapid membrane-initiated and intracellular signaling cascades in POMC and NPY/AgRP neurons to regulate energy homeostasis.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Oline K Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA; Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Martin J Kelly
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
18
|
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
|
19
|
Estrogens synthesized and acting within a spinal oligomer suppress spinal endomorphin 2 antinociception: ebb and flow over the rat reproductive cycle. Pain 2018; 158:1903-1914. [PMID: 28902684 DOI: 10.1097/j.pain.0000000000000991] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The magnitude of antinociception elicited by intrathecal endomorphin 2 (EM2), an endogenous mu-opioid receptor (MOR) ligand, varies across the rat estrous cycle. We now report that phasic changes in analgesic responsiveness to spinal EM2 result from plastic interactions within a novel membrane-bound oligomer containing estrogen receptors (mERs), aromatase (aka estrogen synthase), metabotropic glutamate receptor 1 (mGluR1), and MOR. During diestrus, spinal mERs, activated by locally synthesized estrogens, act with mGluR1 to suppress spinal EM2/MOR antinociception. The emergence of robust spinal EM2 antinociception during proestrus results from the loss of mER-mGluR1 suppression, a consequence of altered interactions within the oligomer. The chemical pairing of aromatase with mERs within the oligomer containing MOR and mGluR1 allows estrogens to function as intracellular messengers whose synthesis and actions are confined to the same signaling oligomer. This form of estrogenic signaling, which we term "oligocrine," enables discrete, highly compartmentalized estrogen/mER-mGluR1 signaling to regulate MOR-mediated antinociception induced by EM2. Finally, spinal neurons were observed not only to coexpress MOR, mERα, aromatase, and mGluR1 but also be apposed by EM2 varicosities. This suggests that modulation of spinal analgesic responsiveness to exogenous EM2 likely reflects changes in its endogenous analgesic activity. Analogous suppression of spinal EM2 antinociception in women (eg, around menses, comparable with diestrus in rats) as well as the (pathological) inability to transition out of that suppressed state at other menstrual cycle stages could underlie, at least in part, the much greater prevalence and severity of chronic pain in women than men.
Collapse
|
20
|
Rivera HM, Stincic TL. Estradiol and the control of feeding behavior. Steroids 2018; 133:44-52. [PMID: 29180290 PMCID: PMC5864536 DOI: 10.1016/j.steroids.2017.11.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022]
Abstract
This review lays out the evidence for the role of E2 in homeostatic and hedonic feeding across several species. While significant effort has been expended on homeostatic feeding research, more studies for hedonic feeding need to be conducted (i.e. are there increases in meal size and enhanced motivation to natural food rewards). By identifying the underlying neural circuitry involved, one can better delineate the mechanisms by which E2 influences feeding behavior. By utilizing more selective neural targeting techniques, such as optogenetics, significant progress can be made toward this goal. Together, behavioral and physiological techniques will help us to better understand neural deficits that can increase the risk for obesity in the absence of E2 (menopause) and aid in developing therapeutic strategies.
Collapse
Affiliation(s)
- H M Rivera
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - T L Stincic
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
21
|
Inhibitor of Differentiation-3 and Estrogenic Endocrine Disruptors: Implications for Susceptibility to Obesity and Metabolic Disorders. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6821601. [PMID: 29507860 PMCID: PMC5817379 DOI: 10.1155/2018/6821601] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 11/07/2017] [Accepted: 11/23/2017] [Indexed: 12/28/2022]
Abstract
The rising global incidence of obesity cannot be fully explained within the context of traditional risk factors such as an unhealthy diet, physical inactivity, aging, or genetics. Adipose tissue is an endocrine as well as a metabolic organ that may be susceptible to disruption by environmental estrogenic chemicals. Since some of the endocrine disruptors are lipophilic chemicals with long half-lives, they tend to bioaccumulate in the adipose tissue of exposed populations. Elevated exposure to these chemicals may predispose susceptible individuals to weight gain by increasing the number and size of fat cells. Genetic studies have demonstrated that the transcriptional regulator inhibitor of differentiation-3 (ID3) promotes high fat diet-induced obesity in vivo. We have shown previously that PCB153 and natural estrogen 17β-estradiol increase ID3 expression. Based on our findings, we postulate that ID3 is a molecular target of estrogenic endocrine disruptors (EEDs) in the adipose tissue and a better understanding of this relationship may help to explain how EEDs can lead to the transcriptional programming of deviant fat cells. This review will discuss the current understanding of ID3 in excess fat accumulation and the potential for EEDs to influence susceptibility to obesity or metabolic disorders via ID3 signaling.
Collapse
|
22
|
Acconcia F, Fiocchetti M, Marino M. Xenoestrogen regulation of ERα/ERβ balance in hormone-associated cancers. Mol Cell Endocrinol 2017; 457:3-12. [PMID: 27816767 DOI: 10.1016/j.mce.2016.10.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/31/2016] [Accepted: 10/31/2016] [Indexed: 02/07/2023]
Abstract
The hormone 17β-estradiol (E2) contributes to body homeostasis maintenance by regulating many different physiological functions in both male and female organs. E2 actions in reproductive and non-reproductive tissues rely on a complex net of nuclear and extra-nuclear signal transduction pathways triggered by at least two estrogen receptor subtypes (ERα and ERβ). Consequently, the de-regulation of E2:ER signaling contributes to the pathogenesis of many diseases including cancer. Among other factors, the ERα/ERβ ratio is considered one of the pivotal mechanisms at the root of E2 action in cancer progression. Remarkably, several natural or synthetic exogenous chemicals, collectively called xenoestrogens, bind to ERs and interfere with their signals and intracellular functions. In this review, the molecular mechanism(s) through which xenoestrogens influence ERα and ERβ intracellular concentrations and the consequences of this influence on E2-related cancer will be discussed.
Collapse
Affiliation(s)
- Filippo Acconcia
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Marco Fiocchetti
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Maria Marino
- Department of Sciences, Section Biomedical Sciences and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy.
| |
Collapse
|
23
|
Kim JY, Park JH, Kim MI, Lee HH, Kim HL, Jeong KS, Moon SO, Kang PW, Park KW, Lee YH, Chun BW. Identification of female-specific blood stains using a 17β-estradiol-targeted aptamer-based sensor. Int J Legal Med 2017; 132:91-98. [PMID: 29086052 DOI: 10.1007/s00414-017-1718-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/11/2017] [Indexed: 01/08/2023]
Abstract
Blood stain evidence obtained from a violent crime scene provides decisive clues that can enable a case to be solved through forensic analyses such as genetic identification. However, collected samples usually contain a mixture of biological material from different sources, making genetic identification difficult. To address this issue, we developed an activatable aptamer sensor targeting 17β-estradiol for detection of female-specific blood in mixed samples. With the sensor, we were able to detect blood originating from females using a variable light source (495 nm). The sensor was especially sensitive to blood from young females (10-40 years) but not to blood from older females (≥ 50 years). Genomic DNA was extracted from the female blood specimens identified by this method and used for quantification and short tandem repeat genotyping. We confirmed that there was no fluorescence interference from the aptamer sensor. These results indicate that this novel aptamer sensor can be used to analyze evidentiary blood samples and thereby facilitate subsequent genetic identification.
Collapse
Affiliation(s)
- Joo-Young Kim
- Crime-scene DNA Section, National Forensic Service, Gwangju Institute, Gwangju, 57231, Republic of Korea
| | - Jung-Hyun Park
- Mibyeong Research Center, Korea Institute of Oriental Medicine, Daejeon, 34054, Republic of Korea
| | - Man Il Kim
- Crime-scene DNA Section, National Forensic Service, Gwangju Institute, Gwangju, 57231, Republic of Korea
| | - Hye Hyeon Lee
- Crime-scene DNA Section, National Forensic Service, Gwangju Institute, Gwangju, 57231, Republic of Korea
| | - Hye Lim Kim
- Crime-scene DNA Section, National Forensic Service, Gwangju Institute, Gwangju, 57231, Republic of Korea
| | - Kyu-Sik Jeong
- Forensic DNA Division, National Forensic Service, Wonju, 26460, Republic of Korea
| | - Sang-Ok Moon
- Crime-scene DNA Section, National Forensic Service, Gwangju Institute, Gwangju, 57231, Republic of Korea
| | - Pil-Won Kang
- Crime-scene DNA Section, National Forensic Service, Gwangju Institute, Gwangju, 57231, Republic of Korea
| | - Ki-Won Park
- Forensic DNA Division, National Forensic Service, Wonju, 26460, Republic of Korea
| | - Yang-Han Lee
- Forensic DNA Division, National Forensic Service, Wonju, 26460, Republic of Korea
| | - Byung-Won Chun
- Forensic DNA Division, National Forensic Service, Wonju, 26460, Republic of Korea.
| |
Collapse
|
24
|
Campello RS, Fátima LA, Barreto-Andrade JN, Lucas TF, Mori RC, Porto CS, Machado UF. Estradiol-induced regulation of GLUT4 in 3T3-L1 cells: involvement of ESR1 and AKT activation. J Mol Endocrinol 2017; 59:257-268. [PMID: 28729437 DOI: 10.1530/jme-17-0041] [Citation(s) in RCA: 24] [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: 06/07/2017] [Accepted: 07/20/2017] [Indexed: 12/11/2022]
Abstract
Impaired insulin-stimulated glucose uptake involves reduced expression of the GLUT4 (solute carrier family 2 facilitated glucose transporter member 4, SLC2A4 gene). 17β-estradiol (E2) modulates SLC2A4/GLUT4 expression, but the involved mechanisms are unclear. Although E2 exerts biological effects by binding to estrogen receptors 1/2 (ESR1/2), which are nuclear transcriptional factors; extranuclear effects have also been proposed. We hypothesize that E2 regulates GLUT4 through an extranuclear ESR1 mechanism. Thus, we investigated the effects of E2 upon (1) subcellular distribution of ESRs and the proto-oncogene tyrosine-protein kinases (SRC) involvement; (2) serine/threonine-protein kinase (AKT) activation; (3) Slc2a4/GLUT4 expression and (4) GLUT4 subcellular distribution and glucose uptake in 3T3-L1 adipocytes. Differentiated 3T3-L1 adipocytes were cultivated or not with E2 for 24 h, and additionally treated or not with ESR1-selective agonist (PPT), ESR1-selective antagonist (MPP) or selective SRC inhibitor (PP2). Subcellular distribution of ESR1, ESR2 and GLUT4 was analyzed by immunocytochemistry; Slc2a4 mRNA and GLUT4 were quantified by qPCR and Western blotting, respectively; plasma membrane GLUT4 translocation and glucose uptake were analyzed under insulin stimulus for 20 min or not. E2 induced (1) translocation of ESR1, but not of ESR2, from nucleus to plasma membrane and AKT phosphorylation, effects mimicked by PPT and blocked by MPP and PP2; (2) increased Slc2a4/GLUT4 expression and (3) increased insulin-stimulated GLUT4 translocation and glucose uptake. In conclusion, E2 treatment promoted a SRC-mediated nucleus-plasma membrane shuttle of ESR1, and increased AKT phosphorylation, Slc2a4/GLUT4 expression and plasma membrane GLUT4 translocation; consequently, improving insulin-stimulated glucose uptake. These results unravel mechanisms through which estrogen improves insulin sensitivity.
Collapse
Affiliation(s)
- Raquel S Campello
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luciana A Fátima
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - João Nilton Barreto-Andrade
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thais F Lucas
- Section of Experimental EndocrinologyDepartment of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Rosana C Mori
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Catarina S Porto
- Section of Experimental EndocrinologyDepartment of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ubiratan F Machado
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
Micevych PE, Mermelstein PG, Sinchak K. Estradiol Membrane-Initiated Signaling in the Brain Mediates Reproduction. Trends Neurosci 2017; 40:654-666. [PMID: 28969926 DOI: 10.1016/j.tins.2017.09.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/28/2017] [Accepted: 09/10/2017] [Indexed: 12/21/2022]
Abstract
Over the past few years our understanding of estrogen signaling in the brain has expanded rapidly. Estrogens are synthesized in the periphery and in the brain, acting on multiple receptors to regulate gene transcription, neural function, and behavior. Various estrogen-sensitive signaling pathways often operate in concert within the same cell, increasing the complexity of the system. In females, estrogen concentrations fluctuate over the estrous/menstrual cycle, dynamically modulating estrogen receptor (ER) expression, activity, and trafficking. These dynamic changes influence multiple behaviors but are particularly important for reproduction. Using the female rodent model, we review our current understanding of estradiol signaling in the regulation of sexual receptivity.
Collapse
Affiliation(s)
- Paul E Micevych
- Department of Neurobiology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), and Laboratory of Neuroendocrinology of the UCLA Brain Research Institute, Los Angeles, CA 90095, USA.
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA 90840, USA
| |
Collapse
|
26
|
Abstract
The hypothalamus is most often associated with innate behaviors such as is hunger, thirst and sex. While the expression of these behaviors important for survival of the individual or the species is nested within the hypothalamus, the desire (i.e., motivation) for them is centered within the mesolimbic reward circuitry. In this review, we will use female sexual behavior as a model to examine the interaction of these circuits. We will examine the evidence for a hypothalamic circuit that regulates consummatory aspects of reproductive behavior, i.e., lordosis behavior, a measure of sexual receptivity that involves estradiol membrane-initiated signaling in the arcuate nucleus (ARH), activating β-endorphin projections to the medial preoptic nucleus (MPN), which in turn modulate ventromedial hypothalamic nucleus (VMH) activity-the common output from the hypothalamus. Estradiol modulates not only a series of neuropeptides, transmitters and receptors but induces dendritic spines that are for estrogenic induction of lordosis behavior. Simultaneously, in the nucleus accumbens of the mesolimbic system, the mating experience produces long term changes in dopamine signaling and structure. Sexual experience sensitizes the response of nucleus accumbens neurons to dopamine signaling through the induction of a long lasting early immediate gene. While estrogen alone increases spines in the ARH, sexual experience increases dendritic spine density in the nucleus accumbens. These two circuits appear to converge onto the medial preoptic area where there is a reciprocal influence of motivational circuits on consummatory behavior and vice versa. While it has not been formally demonstrated in the human, such circuitry is generally highly conserved and thus, understanding the anatomy, neurochemistry and physiology can provide useful insight into the motivation for sexual behavior and other innate behaviors in humans.
Collapse
Affiliation(s)
- Paul E Micevych
- Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine at University of California, Los AngelesLos Angeles, CA, United States.,Brain Research Institute, University of California, Los AngelesLos Angeles, CA, United States
| | - Robert L Meisel
- Department of Neuroscience, University of MinnesotaMinneapolis, MN, United States
| |
Collapse
|
27
|
Inhibition of miR-181a protects female mice from transient focal cerebral ischemia by targeting astrocyte estrogen receptor-α. Mol Cell Neurosci 2017; 82:118-125. [PMID: 28522364 DOI: 10.1016/j.mcn.2017.05.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/03/2017] [Accepted: 05/08/2017] [Indexed: 02/03/2023] Open
Abstract
Whether the effect of miR-181a is sexually dimorphic in stroke is unknown. Prior work showed protection of male mice with miR-181a inhibition. Estrogen receptor-α (ERα) is an identified target of miR181 in endometrium. Therefore we investigated the separate and joint effects of miR-181a inhibition and 17β-estradiol (E2) replacement after ovariectomy. Adult female mice were ovariectomized and implanted with an E2- or vehicle-containing capsule for 14d prior to 1h middle cerebral artery occlusion (MCAO). Each group received either miR-181a antagomir or mismatch control by intracerebroventricular injection 24h before MCAO. After MCAO neurologic deficit and infarct volume were assessed. Primary male and female astrocyte cultures were subjected to glucose deprivation with miR-181a inhibitor or transfection control, and E2 or vehicle control, with/without ESRα knockdown with small interfering RNA. Cell death was assessed by propidium iodide staining, and lactate dehydrogenase assay. A miR-181a/ERα target site blocker (TSB), with/without miR-181a mimic, was used to confirm targeting of ERα by miR-181a in astrocytes. Individually, miR-181a inhibition or E2 decreased infarct volume and improved neurologic score in female mice, and protected male and female astrocyte cultures. Combined miR-181a inhibition plus E2 afforded greater protection of female mice and female astrocyte cultures, but not in male astrocyte cultures. MiR-181a inhibition only increased ERα levels in vivo and in female cultures, while ERα knockdown with siRNA increased cell death in both sexes. Treatment with ERα TSB was strongly protective in both sexes. In conclusion, the results of the present study suggest miR-181a inhibition enhances E2-mediated stroke protection in females in part by augmenting ERα production, a mechanism detected in female mice and female astrocytes. Sex differences were observed with combined miR-181a inhibition/E2 treatment, and miR-181a targeting of ERα.
Collapse
|
28
|
Mittelman-Smith MA, Wong AM, Micevych PE. Estrogen and Progesterone Integration in an in vitro Model of RP3V Kisspeptin Neurons. Neuroendocrinology 2017; 106:101-115. [PMID: 28384629 PMCID: PMC5750133 DOI: 10.1159/000471878] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 03/21/2017] [Indexed: 12/18/2022]
Abstract
Positive feedback on gonadotropin release requires not only estrogen but also progesterone to activate neural circuits. In rodents, ovarian estradiol (E2) stimulates progesterone synthesis in hypothalamic astrocytes (neuroP), needed for the luteinizing hormone (LH) surge. Kisspeptin (kiss) neurons are the principal stimulators of gonadotropin-releasing hormone neurons, and disruption of kiss signaling abrogates the LH surge. Similarly, blocking steroid synthesis in the hypothalamus or deleting classical progesterone receptor (PGR) selectively in kiss neurons prevents the LH surge. These results suggest a synergistic action of E2 and progesterone in kiss neurons to affect gonadotropin release. The mHypoA51, immortalized kiss-expressing neuronal cell line derived from adult female mice, is a tractable model for examining integration of steroid signaling underlying estrogen positive feedback. Here, we report that kiss neurons in vitro integrate E2 and progesterone signaling to increase levels of kiss translation and release. mHypoA51 neurons expressed nonclassical membrane progesterone receptors (mPRα and mPRβ) and E2-inducible PGR, required for progesterone-augmentation of E2-induced kiss expression. With astrocyte-conditioned media or in mHypoA51-astrocyte co-culture, neuroP augmented stimulatory effects of E2 on kiss protein. Progesterone activation of classical, membrane-localized PGR led to activation of MAPK and Src kinases. Importantly, progesterone or Src activation induced release of kiss from E2-primed mHypoA51 neurons. Consistent with previous studies, the present results provide compelling evidence that the interaction of E2 and progesterone stimulates kiss expression and release. Further, these results demonstrate a mechanism though which peripheral E2 may prime kiss neurons to respond to neuroP, mediating estrogen positive feedback.
Collapse
|
29
|
Mittelman-Smith MA, Rudolph LM, Mohr MA, Micevych PE. Rodent Models of Non-classical Progesterone Action Regulating Ovulation. Front Endocrinol (Lausanne) 2017; 8:165. [PMID: 28790975 PMCID: PMC5522857 DOI: 10.3389/fendo.2017.00165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/27/2017] [Indexed: 11/13/2022] Open
Abstract
It is becoming clear that steroid hormones act not only by binding to nuclear receptors that associate with specific response elements in the nucleus but also by binding to receptors on the cell membrane. In this newly discovered manner, steroid hormones can initiate intracellular signaling cascades which elicit rapid effects such as release of internal calcium stores and activation of kinases. We have learned much about the translocation and signaling of steroid hormone receptors from investigations into estrogen receptor α, which can be trafficked to, and signal from, the cell membrane. It is now clear that progesterone (P4) can also elicit effects that cannot be exclusively explained by transcriptional changes. Similar to E2 and its receptors, P4 can initiate signaling at the cell membrane, both through progesterone receptor and via a host of newly discovered membrane receptors (e.g., membrane progesterone receptors, progesterone receptor membrane components). This review discusses the parallels between neurotransmitter-like E2 action and the more recently investigated non-classical P4 signaling, in the context of reproductive behaviors in the rodent.
Collapse
Affiliation(s)
- Melinda A. Mittelman-Smith
- Department of Neurobiology, David Geffen School of Medicine at UCLA, The Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
- *Correspondence: Melinda A. Mittelman-Smith,
| | - Lauren M. Rudolph
- Department of Neurobiology, David Geffen School of Medicine at UCLA, The Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
| | - Margaret A. Mohr
- Department of Neurobiology, David Geffen School of Medicine at UCLA, The Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
| | - Paul E. Micevych
- Department of Neurobiology, David Geffen School of Medicine at UCLA, The Laboratory of Neuroendocrinology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
30
|
Leclercq G, Laïos I, Elie-Caille C, Leiber D, Laurent G, Lesniewska E, Tanfin Z, Jacquot Y. ERα dimerization: a key factor for the weak estrogenic activity of an ERα modulator unable to compete with estradiol in binding assays. J Recept Signal Transduct Res 2016; 37:149-166. [PMID: 27400858 DOI: 10.1080/10799893.2016.1203940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Estrothiazine (ESTZ) is a weak estrogen sharing structural similarities with coumestrol. ESTZ failed to compete with [3H]17β-estradiol ([3H]17β-E2) for binding to the estrogen receptor α (ERα), questioning its ability to interact with the receptor. However, detection by atomic force spectroscopy (AFS) of an ESTZ-induced ERα dimerization has eliminated any remaining doubts. The effect of the compound on the proliferation of ERα-positive and negative breast cancer cells confirmed the requirement of the receptor. The efficiency of ESTZ in MCF-7 cells was weak without any potency to modify the proliferation profile of estradiol and coumestrol. Growth enhancement was associated with a proteasomal degradation of ERα without substantial recruitment of LxxLL coactivators. This may be related to an unusual delay between the acquisition by the receptor of an ERE-binding capacity and the subsequent estrogen-dependent transcription. A complementary ability to enhance TPA-induced AP-1 transcription was observed, even at concentrations insufficient to activate the ERα, suggesting a partly independent mechanism. ESTZ also rapidly and transiently activated ERK1/2 likely through membrane estrogenic pathways provoking a reorganization of the actin network. Finally, the systematic absence of biological responses with an ESTZ derivative unable to induce ERα dimerization stresses the importance of this step in the action of the compound, as reported for conventional estrogens. In view of the existence of many other ERα modulators (endocrine disruptors such as, for example, pesticides, environmental contaminants or phytoestrogens) with extremely weak or similar apparent lack of binding ability, our work may appear as pilot investigation for assessing their mechanism of action.
Collapse
Affiliation(s)
- Guy Leclercq
- a Laboratoire J.-C. Heuson de Cancérologie Mammaire , Université Libre de Bruxelles (U.L.B.), Institut Jules Bordet , Brussels , Belgium
| | - Ioanna Laïos
- a Laboratoire J.-C. Heuson de Cancérologie Mammaire , Université Libre de Bruxelles (U.L.B.), Institut Jules Bordet , Brussels , Belgium
| | - Céline Elie-Caille
- b Institut FEMTO-ST, CNRS UMR 6174, Université de Bourgogne Franche-Comté , Besançon , France
| | - Denis Leiber
- c Laboratoire Signalisation et Régulations Cellulaires , Institut de Biochimie et de Biologie Moléculaire et Cellulaire, CNRS UMR 8619, Université Paris-Sud , Orsay Cedex , France.,d INSERM U1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers , Angers , France
| | - Guy Laurent
- e Service d'Histologie et de Cytologie Expérimentale, Faculté de Médecine et de Pharmacie , Université de Mons-Hainaut , Mons , Belgium
| | - Eric Lesniewska
- f ICB, CNRS UMR 6303, Université de Bourgogne Franche-Comté , Dijon , France
| | - Zahra Tanfin
- c Laboratoire Signalisation et Régulations Cellulaires , Institut de Biochimie et de Biologie Moléculaire et Cellulaire, CNRS UMR 8619, Université Paris-Sud , Orsay Cedex , France
| | - Yves Jacquot
- g Département de Chimie, CNRS UMR 7203 LBM , Sorbonne Universités - UPMC Univ Paris 06, Ecole Normale Supérieure, PSL Research University , Paris , France
| |
Collapse
|
31
|
Gonadal hormone modulation of intracellular calcium as a mechanism of neuroprotection. Front Neuroendocrinol 2016; 42:40-52. [PMID: 26930421 DOI: 10.1016/j.yfrne.2016.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/22/2016] [Accepted: 02/26/2016] [Indexed: 12/28/2022]
Abstract
Hormones have wide-ranging effects throughout the nervous system, including the ability interact with and modulate many aspects of intracellular calcium regulation and calcium signaling. Indeed, these interactions specifically may help to explain the often opposing or paradoxical effects of hormones, such as their ability to both promote and prevent neuronal cell death during development, as well as reduce or exacerbate damage following an insult or injury in adulthood. Here, we review the basic mechanisms underlying intracellular calcium regulation-perhaps the most dynamic and flexible of all signaling molecules-and discuss how gonadal hormones might manipulate these mechanisms to coordinate diverse cellular responses and achieve disparate outcomes. Additional future research that specifically addresses questions of sex and hormone effects on calcium signaling at different ages will be critical to understanding hormone-mediated neuroprotection.
Collapse
|
32
|
Mohr MA, Garcia FL, DonCarlos LL, Sisk CL. Neurons and Glial Cells Are Added to the Female Rat Anteroventral Periventricular Nucleus During Puberty. Endocrinology 2016; 157:2393-402. [PMID: 27145006 PMCID: PMC4891785 DOI: 10.1210/en.2015-2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The anteroventral periventricular nucleus (AVPV) orchestrates the neuroendocrine-positive feedback response that triggers ovulation in female rodents. The AVPV is larger and more cell-dense in females than in males, and during puberty, only females develop the capacity to show a positive feedback response. We previously reported a potential new mechanism to explain this female-specific gain of function during puberty, namely a female-biased sex difference in the pubertal addition of new cells to the rat AVPV. Here we first asked whether this sex difference is due to greater cell proliferation and/or survival in females. Female and male rats received the cell birthdate marker 5-bromo-2'-deoxyuridine (BrdU; 200 mg/kg, ip) on postnatal day (P) 30; brains were collected at short and long intervals after BrdU administration to assess cell proliferation and survival, respectively. Overall, females had more BrdU-immunoreactive cells in the AVPV than did males, with no sex differences in the rate of cell attrition over time. Thus, the sex difference in pubertal addition of AVPV cells appears to be due to greater cell proliferation in females. Next, to determine the phenotype of pubertally born AVPV cells, daily BrdU injections were given to female rats on P28-56, and tissue was collected on P77 to assess colocalization of BrdU and markers for mature neurons or glia. Of the pubertally born AVPV cells, approximately 15% differentiated into neurons, approximately 19% into astrocytes, and approximately 23% into microglia. Thus, both neuro- and gliogenesis occur in the pubertal female rat AVPV and potentially contribute to maturation of female reproductive function.
Collapse
Affiliation(s)
- Margaret A Mohr
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Francisca L Garcia
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Lydia L DonCarlos
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Cheryl L Sisk
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| |
Collapse
|
33
|
Micevych PE, Wong AM, Mittelman-Smith MA. Estradiol Membrane-Initiated Signaling and Female Reproduction. Compr Physiol 2016; 5:1211-22. [PMID: 26140715 DOI: 10.1002/cphy.c140056] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The discoveries of rapid, membrane-initiated steroid actions and central nervous system steroidogenesis have changed our understanding of the neuroendocrinology of reproduction. Classical nuclear actions of estradiol and progesterone steroids affecting transcription are essential. However, with the discoveries of membrane-associated steroid receptors, it is becoming clear that estradiol and progesterone have neurotransmitter-like actions activating intracellular events. Ultimately, membrane-initiated actions can influence transcription. Estradiol membrane-initiated signaling (EMS) modulates female sexual receptivity and estrogen feedback regulating the luteinizing hormone (LH) surge. In the arcuate nucleus, EMS activates a lordosis-regulating circuit that extends to the medial preoptic nucleus and subsequently to the ventromedial nucleus (VMH)--the output from the limbic and hypothalamic regions. Here, we discuss how EMS leads to an active inhibition of lordosis behavior. To stimulate ovulation, EMS facilitates astrocyte synthesis of progesterone (neuroP) in the hypothalamus. Regulation of GnRH release driving the LH surge is dependent on estradiol-sensitive kisspeptin (Kiss1) expression in the rostral periventricular nucleus of the third ventricle (RP3V). NeuroP activation of the LH surge depends on Kiss1, but the specifics of signaling have not been well elucidated. RP3V Kiss1 neurons appear to integrate estradiol and progesterone information which feeds back onto GnRH neurons to stimulate the LH surge. In a second population of Kiss1 neurons, estradiol suppresses the surge but maintains tonic LH release, another critical component of the estrous cycle. Together, evidence suggests that regulation of reproduction involves membrane action of steroids, some of which are synthesized in the brain.
Collapse
Affiliation(s)
- Paul E Micevych
- UCLA - David Geffen School of Medicine Los Angeles, California, USA
| | - Angela May Wong
- UCLA - David Geffen School of Medicine Los Angeles, California, USA
| | | |
Collapse
|
34
|
Galea LAM, Frick KM, Hampson E, Sohrabji F, Choleris E. Why estrogens matter for behavior and brain health. Neurosci Biobehav Rev 2016; 76:363-379. [PMID: 27039345 PMCID: PMC5045786 DOI: 10.1016/j.neubiorev.2016.03.024] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 12/22/2022]
Abstract
The National Institutes of Health (NIH) has required the inclusion of women in clinical studies since 1993, which has enhanced our understanding of how biological sex affects certain medical conditions and allowed the development of sex-specific treatment protocols. However, NIH's policy did not previously apply to basic research, and the NIH recently introduced a new policy requiring all new grant applications to explicitly address sex as a biological variable. The policy itself is grounded in the results of numerous investigations in animals and humans illustrating the existence of sex differences in the brain and behavior, and the importance of sex hormones, particularly estrogens, in regulating physiology and behavior. Here, we review findings from our laboratories, and others, demonstrating how estrogens influence brain and behavior in adult females. Research from subjects throughout the adult lifespan on topics ranging from social behavior, learning and memory, to disease risk will be discussed to frame an understanding of why estrogens matter to behavioral neuroscience.
Collapse
Affiliation(s)
- Liisa A M Galea
- Department of Psychology, Centre for Brain Health, University of British Columbia, Vancouver, BC V6T1Z4, Canada.
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Elizabeth Hampson
- Department of Psychology, University of Western Ontario, London, ON N6A 5C2, Canada
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A&M HSC College of Medicine, Bryan, TX 77807, United States
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
35
|
Amengual-Cladera E, Capllonch-Amer G, Lladó I, Gianotti M, Proenza AM. Proteomic study of periovarian adipose tissue in 17β-estradiol-treated and untreated ovariectomized rats. Biochem Cell Biol 2016; 94:167-75. [PMID: 26914441 DOI: 10.1139/bcb-2015-0077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Taking into account the sexual dimorphism previously found in white adipose tissue (WAT) regarding mitochondrial function and biogenesis, as well as insulin sensitivity, the aim of this study was to go further into the role of sex hormones in this dimorphism. To achieve this objective, we used ovariectomized rats and performed a screening by means of proteomic analyses of the periovarian WAT, combined with a study of the protein levels of specific factors involved in mitochondrial function. Rats were ovariectomized at 5 weeks of age and subcutaneously injected every 48 h with corn-oil (OVX group) or with 17β-estradiol (E2, 10 μg/kg body mass; OVX + E2 group) for 4 weeks prior to sacrifice. Beside proteomic analysis, protein levels of Transcription Factor A, Mitochondrial (TFAM), cytochrome oxidase (COX)II, and COXIV were determined by Western blot, and mRNA levels of peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α, ERα, ERβ, lipoprotein lipase (LPL), peroxisome proliferator-activated receptor-γ (PPARγ), and adiponectin were quantified by real-time PCR. Our results show that ovariectomy leads to an increase in anabolic processes and inflammatory protein levels as well as to a decrease in some of the markers of mitochondrial function, which are restored, at least in part, by E2 supplementation. Indeed, this E2 supplementation seems to be counteracted by a decline in ERα and in the ERα to ERβ ratio values that could be directed to avoid an over-stimulation of the E2 signaling pathway, given the possibility of an activation of extra-gonadal steroid biosynthetic pathways.
Collapse
Affiliation(s)
- Emilia Amengual-Cladera
- a Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Palma de Mallorca, Spain.,b Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, C/Sinesio Delgado 4, 28029 Madrid, Spain
| | - Gabriela Capllonch-Amer
- a Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Palma de Mallorca, Spain
| | - Isabel Lladó
- a Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Palma de Mallorca, Spain.,b Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, C/Sinesio Delgado 4, 28029 Madrid, Spain
| | - Magdalena Gianotti
- a Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Palma de Mallorca, Spain.,b Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, C/Sinesio Delgado 4, 28029 Madrid, Spain
| | - Ana M Proenza
- a Grup Metabolisme Energètic i Nutrició, Departament de Biologia Fonamental i Ciències de la Salut, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Palma de Mallorca, Spain.,b Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, C/Sinesio Delgado 4, 28029 Madrid, Spain
| |
Collapse
|
36
|
Bowlby DA, Brown TJ, Hochberg RB, MacLusky NJ. In vitro Autoradiographic Analysis of Regional Changes in Estrogen Receptor Alpha in the Brains of Cycling Female Rats. Neuroendocrinology 2016; 103:538-51. [PMID: 26422138 DOI: 10.1159/000441077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/15/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS The contributions of the three principal ovarian steroid hormones (estradiol, progesterone and testosterone) to the regulation of estrogen receptor alpha (ERα) levels in the rat brain were examined during the estrous cycle. METHODS Receptor concentrations were measured using an in vitro autoradiographic technique designed to separately quantify free, unoccupied receptors and receptors 'occupied' by (bound to) endogenous hormone. RESULTS ERα occupation increased at proestrus and declined at estrus, reflecting changes in circulating estradiol and testosterone levels. Total ERα content followed a pattern that was the inverse of the occupation data, falling over the night of proestrus. Between 2.00 and 10.00 a.m. on the day of estrus, total ERα concentrations recovered in all brain regions except the ventromedial nucleus (VMN), in which ERα binding remained depressed at estrus. Administration of the progesterone antagonist mifepristone on the afternoon of proestrus resulted in recovery of ERα levels in the VMN by the morning of estrus, consistent with the hypothesis that the preovulatory progesterone surge selectively inhibits VMN ERα expression. Residual ERα occupation observed at estrus, when estradiol is not detectable in the serum, likely reflects intracranial aromatization of circulating androgens, since the pattern of receptor occupation observed at this stage of the cycle could be reproduced in ovariectomized rats by replacement with testosterone. CONCLUSION These findings indicate that ERα binding in the brain fluctuates during the rat estrous cycle in a region-specific manner and suggest that local aromatization of testosterone may contribute significantly to ERα occupation when circulating estradiol levels are low.
Collapse
Affiliation(s)
- Deborah A Bowlby
- Department of Biomedical Sciences, University of Guelph, Guelph, Ont., Canada
| | | | | | | |
Collapse
|
37
|
Clark S, Pollard K, Rainville J, Vasudevan N. Detection of the Phosphorylation of the Estrogen Receptor α as an Outcome of GPR30 Activation. Methods Mol Biol 2016; 1366:457-470. [PMID: 26585157 DOI: 10.1007/978-1-4939-3127-9_36] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Phosphorylation of the serine residues in estrogen receptor (ER) α is important in transcriptional activation. Hence, methods to detect such posttranslational modification events are valuable. We describe, in detail, the analysis of the phosphorylated ERα by electrophoretic separation of proteins and subsequent immuno-blotting techniques. In particular, phosphorylation of the ERα is one possible outcome of activation of the putative membrane estrogen receptor (mER), GPR30. Hence, phosphorylation represents a cross talk event between GPR30 and ERα and may be important in estrogen-regulated physiology.
Collapse
Affiliation(s)
- Sara Clark
- Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA, 70118, USA
| | - Kevin Pollard
- The Neuroscience Program, Tulane University, New Orleans, LA, 70118, USA
| | - Jennifer Rainville
- Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA, 70118, USA
| | - Nandini Vasudevan
- Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA, 70118, USA.
- The Neuroscience Program, Tulane University, New Orleans, LA, 70118, USA.
| |
Collapse
|
38
|
Na W, Park JW, An JH, Jang J. Size-controllable ultrathin carboxylated polypyrrole nanotube transducer for extremely sensitive 17β-estradiol FET-type biosensors. J Mater Chem B 2016; 4:5025-5034. [DOI: 10.1039/c6tb00897f] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Size-controllable aptamer conjugated ultrathin carboxylated polypyrrole nanotubes (A-UCPPyNTs) were successfully fabricated as transducers in 17β-estradiol field-effect transistor (FET)-type biosensors which has extremely high sensitivity (∼1 fM) and unique selectivity.
Collapse
Affiliation(s)
- Wonjoo Na
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Jin Wook Park
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Ji Hyun An
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Jyongsik Jang
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| |
Collapse
|
39
|
Application of Circular Dichroism Spectroscopy to the Analysis of the Interaction Between the Estrogen Receptor Alpha and Coactivators: The Case of Calmodulin. Methods Mol Biol 2015; 1366:241-259. [PMID: 26585140 DOI: 10.1007/978-1-4939-3127-9_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The estrogen receptor α ligand-binding domain (ERα-LBD) binds the natural hormone 17β-estradiol (E2) to induce transcription and cell proliferation. This process occurs with the contribution of protein and peptide partners (also called coactivators) that can modulate the structure of ERα, and therefore its specificity of action. As with most transcription factors, ERα exhibits a high content of α helix, making it difficult to routinely run spectroscopic studies capable of deciphering the secondary structure of the different partners under binding conditions. Ca(2+)-calmodulin, a protein also highly structured in α-helix, is a key coactivator for ERα activity. Here, we show how circular dichroism can be used to study the interaction of ERα with Ca(2+)-calmodulin. Our approach allows the determination not only of the conformational changes induced upon complex formation but also the dissociation constant (K d) of this interaction.
Collapse
|
40
|
Frick KM, Kim J, Tuscher JJ, Fortress AM. Sex steroid hormones matter for learning and memory: estrogenic regulation of hippocampal function in male and female rodents. Learn Mem 2015; 22:472-93. [PMID: 26286657 PMCID: PMC4561402 DOI: 10.1101/lm.037267.114] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/09/2015] [Indexed: 01/24/2023]
Abstract
Ample evidence has demonstrated that sex steroid hormones, such as the potent estrogen 17β-estradiol (E2), affect hippocampal morphology, plasticity, and memory in male and female rodents. Yet relatively few investigators who work with male subjects consider the effects of these hormones on learning and memory. This review describes the effects of E2 on hippocampal spinogenesis, neurogenesis, physiology, and memory, with particular attention paid to the effects of E2 in male rodents. The estrogen receptors, cell-signaling pathways, and epigenetic processes necessary for E2 to enhance memory in female rodents are also discussed in detail. Finally, practical considerations for working with female rodents are described for those investigators thinking of adding females to their experimental designs.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| |
Collapse
|
41
|
Lacreuse A, Mong JA, Hara Y. Neurocognitive effects of estrogens across the adult lifespan in nonhuman primates: State of knowledge and new perspectives. Horm Behav 2015; 74:157-66. [PMID: 25762288 DOI: 10.1016/j.yhbeh.2015.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 02/26/2015] [Accepted: 03/02/2015] [Indexed: 01/29/2023]
Abstract
This article is part of a Special Issue "Estradiol and cognition". This review discusses the unique contribution of nonhuman primate research to our understanding of the neurocognitive effects of estrogens throughout the adult lifespan in females. Mounting evidence indicates that estrogens affect many aspects of hippocampal, prefrontal and cholinergic function in the primate brain and the underlying mechanisms are beginning to be elucidated. In addition, estrogens may also influence cognitive function indirectly, via the modulation of other systems that impact cognition. We will focus on the effects of estrogens on sleep and emphasize the need for primate models to better understand these complex interactions. Continued research with nonhuman primates is essential for the development of therapies that are optimal for the maintenance of women's cognitive health throughout the lifespan.
Collapse
Affiliation(s)
- Agnès Lacreuse
- Department of Psychological and Brain Sciences, University of Massachusetts at Amherst, MA, USA.
| | - Jessica A Mong
- Department of Pharmacology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Yuko Hara
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
42
|
Hara Y, Waters EM, McEwen BS, Morrison JH. Estrogen Effects on Cognitive and Synaptic Health Over the Lifecourse. Physiol Rev 2015; 95:785-807. [PMID: 26109339 PMCID: PMC4491541 DOI: 10.1152/physrev.00036.2014] [Citation(s) in RCA: 283] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Estrogen facilitates higher cognitive functions by exerting effects on brain regions such as the prefrontal cortex and hippocampus. Estrogen induces spinogenesis and synaptogenesis in these two brain regions and also initiates a complex set of signal transduction pathways via estrogen receptors (ERs). Along with the classical genomic effects mediated by activation of ER α and ER β, there are membrane-bound ER α, ER β, and G protein-coupled estrogen receptor 1 (GPER1) that can mediate rapid nongenomic effects. All key ERs present throughout the body are also present in synapses of the hippocampus and prefrontal cortex. This review summarizes estrogen actions in the brain from the standpoint of their effects on synapse structure and function, noting also the synergistic role of progesterone. We first begin with a review of ER subtypes in the brain and how their abundance and distributions are altered with aging and estrogen loss (e.g., ovariectomy or menopause) in the rodent, monkey, and human brain. As there is much evidence that estrogen loss induced by menopause can exacerbate the effects of aging on cognitive functions, we then review the clinical trials of hormone replacement therapies and their effectiveness on cognitive symptoms experienced by women. Finally, we summarize studies carried out in nonhuman primate models of age- and menopause-related cognitive decline that are highly relevant for developing effective interventions for menopausal women. Together, we highlight a new understanding of how estrogen affects higher cognitive functions and synaptic health that go well beyond its effects on reproduction.
Collapse
Affiliation(s)
- Yuko Hara
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - Elizabeth M Waters
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - Bruce S McEwen
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - John H Morrison
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| |
Collapse
|
43
|
Kelly MJ, Rønnekleiv OK. Minireview: neural signaling of estradiol in the hypothalamus. Mol Endocrinol 2015; 29:645-57. [PMID: 25751314 PMCID: PMC4415204 DOI: 10.1210/me.2014-1397] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/02/2015] [Indexed: 12/22/2022] Open
Affiliation(s)
- Martin J Kelly
- Departments of Physiology and Pharmacology (M.J.K., O.K.R.) and Anesthesiology and Perioperative Medicine (O.K.R.), Oregon Health and Science University, Portland, Oregon 97239; and Division of Neuroscience (M.J.K., O.K.R.), Oregon National Primate Research Center; Oregon Health and Science University, Beaverton, Oregon 97006
| | | |
Collapse
|
44
|
Tan Y, Wei T. Detection of 17β-estradiol in water samples by a novel double-layer molecularly imprinted film-based biosensor. Talanta 2015; 141:279-87. [PMID: 25966415 DOI: 10.1016/j.talanta.2015.04.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/02/2015] [Accepted: 04/05/2015] [Indexed: 10/23/2022]
Abstract
This study reports a novel double-layer molecularly imprinted film (MIF)-based biosensor for rapid, sensitive and highly selective detection of small molecule 17β-estradiol (E2) that is frequently detected in environmental water samples. In this system, the modification of gold surface of SPR chip was performed by 1-dodecanethiol. Then double-layer MIF was generated on the 1-dodecanethiol modified gold surface. The non-modified and imprinted surfaces were characterized by atomic force microscopy (AFM), scanning electron microscope (SEM) and contact angle measurements. Analysis of surface plasmon resonance (SPR) spectroscopy showed that the imprinted sensing film displayed good selectivity for E2 compared to other analog molecules and NIF. A good linear relationship was obtained between the SPR angle and E2 concentrations over a range of 2.50×10(-13)-2.50×10(-)(9)mol/L (R(2)=0.993) with the lowest measurable concentration of 2.50×10(-13)mol/L. The sensor can be regenerated with the mixture of acetic acid and PBS buffer (v/v=1:9) as a desorption agent over tens of times without significant deterioration of the sensor performance. Potential interference of real environmental sample matrix was assessed by spiked samples in several waste seawater effluents. This portable sensor system can be successfully applied for on-site real-time inexpensive and easy-to-use monitoring of E2 or other small molecule pollutants in environmental samples such as effluents or water bodies.
Collapse
Affiliation(s)
- Yuan Tan
- Key Laboratory of Cluster Science of Ministry of Education, Beijing Institute of Technology, Beijing 100081, People's Republic of China
| | - Tianxin Wei
- Key Laboratory of Cluster Science of Ministry of Education, Beijing Institute of Technology, Beijing 100081, People's Republic of China.
| |
Collapse
|
45
|
Totta P, Pesiri V, Enari M, Marino M, Acconcia F. Clathrin Heavy Chain Interacts With Estrogen Receptor α and Modulates 17β-Estradiol Signaling. Mol Endocrinol 2015; 29:739-55. [PMID: 25860340 DOI: 10.1210/me.2014-1385] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
17β-estradiol (E2)-induced signaling and control of estrogen receptor (ER)α degradation both play a major role in breast cancer cell proliferation. We recently reported the involvement of lysosomal function in both E2-dependent ERα breakdown and E2-induced cell proliferation and thus hypothesized a role for endocytic proteins in ERα signaling. An small interfering RNA screen identified proteins that regulate intracellular endocytic traffic and whose silencing alters E2-induced ERα degradation. One such protein was the clathrin heavy chain (CHC), whose role in E2:ERα signaling to cell proliferation is unknown. Here, we show that CHC physically interacts with ERα in the cytoplasm of breast cancer cells and regulates E2-induced cell proliferation. Surprisingly, the CHC:ERα interaction is required to sustain E2 signaling but is dispensable for ERα degradation. Our data also demonstrate that many membrane trafficking proteins contribute to the regulation of ERα degradation, thus unraveling the contribution of endocytic proteins in E2:ERα signaling.
Collapse
Affiliation(s)
- Pierangela Totta
- Department of Sciences (P.T., V.P., M.M., F.A.), Section Biomedical Sciences and Technology, University Roma Tre, I-00146, Rome, Italy; and Division of Refractory Cancer Research (M.E.), National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | |
Collapse
|
46
|
Wong AM, Abrams MC, Micevych PE. β-arrestin regulates estradiol membrane-initiated signaling in hypothalamic neurons. PLoS One 2015; 10:e0120530. [PMID: 25803606 PMCID: PMC4372564 DOI: 10.1371/journal.pone.0120530] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/23/2015] [Indexed: 12/26/2022] Open
Abstract
Estradiol (E2) action in the nervous system is the result of both direct nuclear and membrane-initiated signaling (EMS). E2 regulates membrane estrogen receptor-α (ERα) levels through opposing mechanisms of EMS-mediated trafficking and internalization. While ß-arrestin-mediated mERα internalization has been described in the cortex, a role of ß-arrestin in EMS, which underlies multiple physiological processes, remains undefined. In the arcuate nucleus of the hypothalamus (ARH), membrane-initiated E2 signaling modulates lordosis behavior, a measure of female sexually receptivity. To better understand EMS and regulation of ERα membrane levels, we examined the role of ß-arrestin, a molecule associated with internalization following agonist stimulation. In the present study, we used an immortalized neuronal cell line derived from embryonic hypothalamic neurons, the N-38 line, to examine whether ß-arrestins mediate internalization of mERα. β-arrestin-1 (Arrb1) was found in the ARH and in N-38 neurons. In vitro, E2 increased trafficking and internalization of full-length ERα and ERαΔ4, an alternatively spliced isoform of ERα, which predominates in the membrane. Treatment with E2 also increased phosphorylation of extracellular-signal regulated kinases 1/2 (ERK1/2) in N-38 neurons. Arrb1 siRNA knockdown prevented E2-induced ERαΔ4 internalization and ERK1/2 phosphorylation. In vivo, microinfusions of Arrb1 antisense oligodeoxynucleotides (ODN) into female rat ARH knocked down Arrb1 and prevented estradiol benzoate-induced lordosis behavior compared with nonsense scrambled ODN (lordosis quotient: 3 ± 2.1 vs. 85.0 ± 6.0; p < 0.0001). These results indicate a role for Arrb1 in both EMS and internalization of mERα, which are required for the E2-induction of female sexual receptivity.
Collapse
Affiliation(s)
- Angela M. Wong
- Department of Neurobiology David Geffen School of Medicine at UCLA and Laboratory of Neuroendocrinology of the Brain Research Institute, at University of California Los Angeles, Los Angeles, California, United States of America
| | - Matthew C. Abrams
- Department of Neurobiology David Geffen School of Medicine at UCLA and Laboratory of Neuroendocrinology of the Brain Research Institute, at University of California Los Angeles, Los Angeles, California, United States of America
| | - Paul E. Micevych
- Department of Neurobiology David Geffen School of Medicine at UCLA and Laboratory of Neuroendocrinology of the Brain Research Institute, at University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Sinchak K, Dalhousay L, Sanathara N. Orphanin FQ-ORL-1 regulation of reproduction and reproductive behavior in the female. VITAMINS AND HORMONES 2015; 97:187-221. [PMID: 25677773 DOI: 10.1016/bs.vh.2014.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Orphanin FQ (OFQ/N) and its receptor, opioid receptor-like receptor-1 (ORL-1), are expressed throughout steroid-responsive limbic and hypothalamic circuits that regulate female ovarian hormone feedback and reproductive behavior circuits. The arcuate nucleus of the hypothalamus (ARH) is a brain region that expresses OFQ/N and ORL-1 important for both sexual behavior and modulating estradiol feedback loops. Within the ARH, the activation of the OFQ/N-ORL-1 system facilitates sexual receptivity (lordosis) through the inhibition of β-endorphin neuronal activity. Estradiol initially activates ARH β-endorphin neurons to inhibit lordosis. Simultaneously, estradiol upregulates coexpression of OFQ/N and progesterone receptors and ORL-1 in ARH β-endorphin neurons. Ovarian hormones regulate pre- and postsynaptic coupling of ORL-1 to its G protein-coupled signaling pathways. When the steroid-primed rat is nonreceptive, estradiol acts pre- and postsynaptically to decrease the ability of the OFQ/N-ORL-1 system to inhibit ARH β-endorphin neurotransmission. Conversely, when sexually receptive, ORL-1 signaling is restored to inhibit β-endorphin neurotransmission. Although steroid signaling that facilitates lordosis converges to deactivate ARH β-endorphin neurons, estradiol-only facilitation of lordosis requires the activation of ORL-1, but estradiol+progesterone does not, indicating that multiple circuits mediate ovarian hormone signaling to deactivate ARH β-endorphin neurons. Research on the role of OFQ/N-ORL-1 in ovarian hormone feedback loops is just beginning. In the rat, OFQ/N may act to terminate gonadotropin-releasing hormone and luteinizing hormone release under positive and negative feedbacks. In the ewe, it appears to directly inhibit gonadotropin-releasing hormone release to mediate progesterone-negative feedback. As a whole, the localization and actions of OFQ/N-ORL-1 system indicate that it may mediate the actions of estradiol and progesterone to synchronize reproductive behavior and ovarian hormone feedback loops.
Collapse
Affiliation(s)
- Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, California, USA.
| | - Lauren Dalhousay
- Department of Biological Sciences, California State University, Long Beach, California, USA
| | - Nayna Sanathara
- Department of Pharmacological Sciences, University of California, Irvine, California, USA
| |
Collapse
|
48
|
Kumar A, Storman EM, Liu NJ, Gintzler AR. Estrogens Suppress Spinal Endomorphin 2 Release in Female Rats in Phase with the Estrous Cycle. Neuroendocrinology 2015; 102:33-43. [PMID: 25925013 PMCID: PMC4575620 DOI: 10.1159/000430817] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/20/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND/AIMS Male and female rats differ in their ability to utilize spinal endomorphin 2 (EM2; the predominant mu-opioid receptor ligand in spinal cord) and in the mechanisms that underlie spinal EM2 analgesic responsiveness. We investigated the relevance of spinal estrogen receptors (ERs) to the in vivo regulation of spinal EM2 release. METHODS ER antagonists were administered directly to the lumbosacral spinal cord of male and female rats, intrathecal perfusate was collected, and resulting changes in EM2 release were quantified using a plate-based radioimmunoassay. RESULTS Intrathecal application of an antagonist of either estrogen receptor-α (ERα) or the ER GPR30 failed to alter spinal EM2 release. Strikingly, however, the concomitant blockade of ERα and GPR30 enhanced spinal EM2 release. This effect was sexually dimorphic, being absent in males. Furthermore, the magnitude of the enhancement of spinal EM2 release in females was dependent upon estrous cycle stage, suggesting a relationship with circulating levels of 17β-estradiol. The rapid onset of enhanced EM2 release following intrathecal application of ERα/GPR30 antagonists (within 30-40 min) suggests mediation via ERs in the plasma membrane, not the nucleus. Notably, both ovarian and spinally synthesized estrogens are essential for membrane ER regulation of spinal EM2 release. CONCLUSION These findings underscore the importance of estrogens for the regulation of spinal EM2 activity and, by extension, endogenous spinal EM2 antinociception in females. Components of the spinal estrogenic mechanism(s) that suppress EM2 release could represent novel drug targets for improving utilization of endogenous spinal EM2, and thereby pain management in women.
Collapse
Affiliation(s)
- Arjun Kumar
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, N.Y., USA
| | | | | | | |
Collapse
|
49
|
Rainville J, Pollard K, Vasudevan N. Membrane-initiated non-genomic signaling by estrogens in the hypothalamus: cross-talk with glucocorticoids with implications for behavior. Front Endocrinol (Lausanne) 2015; 6:18. [PMID: 25762980 PMCID: PMC4329805 DOI: 10.3389/fendo.2015.00018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 01/30/2015] [Indexed: 12/12/2022] Open
Abstract
The estrogen receptor and glucocorticoid receptor are members of the nuclear receptor superfamily that can signal using both non-genomic and genomic transcriptional modes. Though genomic modes of signaling have been well characterized and several behaviors attributed to this signaling mechanism, the physiological significance of non-genomic modes of signaling has not been well understood. This has partly been due to the controversy regarding the identity of the membrane ER (mER) or membrane GR (mGR) that may mediate rapid, non-genomic signaling and the downstream signaling cascades that may result as a consequence of steroid ligands binding the mER or the mGR. Both estrogens and glucocorticoids exert a number of actions on the hypothalamus, including feedback. This review focuses on the various candidates for the mER or mGR in the hypothalamus and the contribution of non-genomic signaling to classical hypothalamically driven behaviors and changes in neuronal morphology. It also attempts to categorize some of the possible functions of non-genomic signaling at both the cellular level and at the organismal level that are relevant for behavior, including some behaviors that are regulated by both estrogens and glucocorticoids in a potentially synergistic manner. Lastly, it attempts to show that steroid signaling via non-genomic modes may provide the organism with rapid behavioral responses to stimuli.
Collapse
Affiliation(s)
- Jennifer Rainville
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
| | - Kevin Pollard
- Neuroscience Program, Tulane University, New Orleans, LA, USA
| | - Nandini Vasudevan
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
- Neuroscience Program, Tulane University, New Orleans, LA, USA
- *Correspondence: Nandini Vasudevan, Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA 70118, USA e-mail:
| |
Collapse
|
50
|
Sellers K, Raval P, Srivastava DP. Molecular signature of rapid estrogen regulation of synaptic connectivity and cognition. Front Neuroendocrinol 2015; 36:72-89. [PMID: 25159586 DOI: 10.1016/j.yfrne.2014.08.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/11/2014] [Accepted: 08/14/2014] [Indexed: 12/14/2022]
Abstract
There is now a growing appreciation that estrogens are capable of rapidly activating a number of signaling cascades within the central nervous system. In addition, there are an increasing number of studies reporting that 17β-estradiol, the major biologically active estrogen, can modulate cognition within a rapid time frame. Here we review recent studies that have begun to uncover the molecular and cellular framework which contributes to estrogens ability to rapidly modulate cognition. We first describe the mechanisms by which estrogen receptors (ERs) can couple to intracellular signaling cascades, either directly, or via the transactivation of other receptors. Subsequently, we review the evidence that estrogen can rapidly modulate both neuronal function and structure in the hippocampus and the cortex. Finally, we will discuss how estrogens may influence cognitive function through the modulation of neuronal structure, and the implications this may have on the treatment of a range of brain disorders.
Collapse
Affiliation(s)
- Katherine Sellers
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK.
| |
Collapse
|