101
|
Etgen AM, Jover-Mengual T, Suzanne Zukin R. Neuroprotective actions of estradiol and novel estrogen analogs in ischemia: translational implications. Front Neuroendocrinol 2011; 32:336-52. [PMID: 21163293 PMCID: PMC3080451 DOI: 10.1016/j.yfrne.2010.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/06/2010] [Accepted: 12/09/2010] [Indexed: 11/15/2022]
Abstract
This review highlights our investigations into the neuroprotective efficacy of estradiol and other estrogenic agents in a clinically relevant animal model of transient global ischemia, which causes selective, delayed death of hippocampal CA1 neurons and associated cognitive deficits. We find that estradiol rescues a significant number of CA1 pyramidal neurons that would otherwise die in response to global ischemia, and this is true when hormone is provided as a long-term pretreatment at physiological doses or as an acute treatment at the time of reperfusion. In addition to enhancing neuronal survival, both forms of estradiol treatment induce measurable cognitive benefit in young animals. Moreover, estradiol and estrogen analogs that do not bind classical nuclear estrogen receptors retain their neuroprotective efficacy in middle-aged females deprived of ovarian hormones for a prolonged duration (8weeks). Thus, non-feminizing estrogens may represent a new therapeutic approach for treating the neuronal damage associated with global ischemia.
Collapse
Affiliation(s)
- Anne M. Etgen
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
- Department of Psychiatry & Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| | - Teresa Jover-Mengual
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Valencia, Campus de Burjassot, Avda. Vicent Andrés Estellés, s/n, 46100 Burjassot, Valencia SPAIN
| | - R. Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| |
Collapse
|
102
|
Herrera JL, Fernandez C, Diaz M, Cury D, Marin R. Estradiol and tamoxifen differentially regulate a plasmalemmal voltage-dependent anion channel involved in amyloid-beta induced neurotoxicity. Steroids 2011; 76:840-4. [PMID: 21354436 DOI: 10.1016/j.steroids.2011.02.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 01/09/2011] [Accepted: 02/16/2011] [Indexed: 01/06/2023]
Abstract
There is a wealth of information indicating that estradiol exerts rapid actions involved in neuroprotection and cognitive-enhancing effects. Some of these effects appear to delay onset, or even ameliorate, the neuropathology of Alzheimer's disease (AD), although some controversy exists about the beneficial brain effects of estrogen therapies. Therefore, it is crucial to better understand the mechanisms developed by 17β-estradiol to signal in the brain. At the neuronal membrane, the hormone can rapidly interact with estrogen receptors (mERs) or activate other receptors, such as G protein-coupled and ionotropic receptors. And the list of membrane signalling molecules modulated by estradiol in neurons is increasing. VDAC is a voltage-dependent anion channel, known as a mitochondrial porin which is also found at the neuronal membrane, where it appears to be involved in redox regulation, extrinsic apoptosis and amyloid beta neurotoxicity. Moreover, VDAC is present in neuronal lipid rafts, where it is associated with estrogen receptor α-like (mER), forming part of a macromolecular complex together with caveolin-1 and other signalling proteins related to neuronal preservation. Interestingly, we have recently found that 17β-estradiol rapidly promotes VDAC phosphorylation through the activation of protein kinase A (PKA) and Src-kinase, which may be relevant to maintain this channel inactivated. On the contrary, tamoxifen, a selective estrogen receptor modulator (SERM), provokes the dephosphorylation of VDAC, and eventually its opening, by activating a cascade of phosphatases, including protein phosphatase 2 (PP2A). This review will focus on the relevance of these novel findings in the alternative estrogen mechanisms to achieve neuroprotection related to AD.
Collapse
Affiliation(s)
- Jose Luis Herrera
- Laboratory of Cellular Neurobiology, Department of Physiology & Institute of Biomedical Technologies, University of La Laguna, School of Medicine, Santa Cruz de Tenerife, Spain
| | | | | | | | | |
Collapse
|
103
|
Zheng S, Huang J, Zhou K, Zhang C, Xiang Q, Tan Z, Wang T, Fu X. 17β-Estradiol enhances breast cancer cell motility and invasion via extra-nuclear activation of actin-binding protein ezrin. PLoS One 2011; 6:e22439. [PMID: 21818323 PMCID: PMC3144228 DOI: 10.1371/journal.pone.0022439] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 06/27/2011] [Indexed: 12/21/2022] Open
Abstract
Estrogen promotes breast cancer metastasis. However, the detailed mechanism remains largely unknown. The actin binding protein ezrin is a key component in tumor metastasis and its over-expression is positively correlated to the poor outcome of breast cancer. In this study, we investigate the effects of 17β-estradiol (E2) on the activation of ezrin and its role in estrogen-dependent breast cancer cell movement. In T47-D breast cancer cells, E2 rapidly enhances ezrin phosphorylation at Thr567 in a time- and concentration-dependent manner. The signalling cascade implicated in this action involves estrogen receptor (ER) interaction with the non-receptor tyrosine kinase c-Src, which activates the phosphatidylinositol-3 kinase/Akt pathway and the small GTPase RhoA/Rho-associated kinase (ROCK-2) complex. E2 enhances the horizontal cell migration and invasion of T47-D breast cancer cells in three-dimensional matrices, which is reversed by transfection of cells with specific ezrin siRNAs. In conclusion, E2 promotes breast cancer cell movement and invasion by the activation of ezrin. These results provide novel insights into the effects of estrogen on breast cancer progression and highlight potential targets to treat endocrine-sensitive breast cancers.
Collapse
Affiliation(s)
- Shuhui Zheng
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinghe Huang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kewen Zhou
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chengxi Zhang
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiuling Xiang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi Tan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tinghuai Wang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XDF); (THW)
| | - Xiaodong Fu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XDF); (THW)
| |
Collapse
|
104
|
17Beta-estradiol differentially protects cortical pericontusional zone from programmed cell death after traumatic cerebral contusion at distinct stages via non-genomic and genomic pathways. Mol Cell Neurosci 2011; 48:185-94. [PMID: 21803156 DOI: 10.1016/j.mcn.2011.07.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 06/23/2011] [Accepted: 07/14/2011] [Indexed: 11/20/2022] Open
Abstract
Pericontusional zone (PCZ) of traumatic cerebral contusion is a target of pharmacological intervention. Our previous study indicated that 17beta-estradiol has a protective role in PCZ after traumatic cerebral contusion via the upregulation of estrogen receptor (ER) alpha mRNA induction and protein expression as well as inhibition of caspase-3 activation, suggesting that genomic signaling pathway is implicated in the protective effect of 17beta-estrodiol. Recent findings demonstrated that 17beta-estradiol also acts on the extranuclear/membrane ER to activate non-genomic signaling pathway to regulate cellular functions and exert the protective effect in the brain. It is still unclear how and whether genomic and non-genomic pathways of 17beta-estradiol are involved in the neuroprotection in PCZ. Our current study demonstrates that 17beta-estradiol activates ERK1/2 and Akt at the early stage and induces ERalpha and survivin mRNA at the late stage to modulate its protection via the suppression of caspase-3 activation in PCZ. These findings suggest that 17beta-estrodiol differentially plays its protective roles via genomic and non-genomic signaling pathways in PCZ after traumatic cerebral contusion.
Collapse
|
105
|
Grassi S, Tozzi A, Costa C, Tantucci M, Colcelli E, Scarduzio M, Calabresi P, Pettorossi VE. Neural 17β-estradiol facilitates long-term potentiation in the hippocampal CA1 region. Neuroscience 2011; 192:67-73. [PMID: 21749911 DOI: 10.1016/j.neuroscience.2011.06.078] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/23/2011] [Accepted: 06/28/2011] [Indexed: 10/18/2022]
Abstract
In the hippocampal formation many neuromodulators are possibly implied in the synaptic plasticity such as the long-term potentiation (LTP) induced by high-frequency stimulation (HFS) of afferent fibers. We investigated the involvement of locally synthesized neural 17β-estradiol (nE(2)) in the induction of HFS-LTP in hippocampal slices from male rats by stimulating the Schaffer collateral fibers and recording the evoked field excitatory postsynaptic potential (fEPSP) in the CA1 region. We demonstrated that either the blockade of nE(2) synthesis by the aromatase inhibitor letrozole, or the antagonism of E(2) receptors (ERs) by ICI 182,780 did not prevent the induction of HFS-LTP, but reduced its amplitude by ∼60%, without influencing its maintenance. Moreover, letrozole and ICI 182,780 did not affect the first short-term post-tetanic component of LTP and the paired-pulse facilitation (PPF). These findings demonstrate that nE(2) plays an important role in the induction phase of HFS-dependent LTP. Since the basal responses were not affected by the blocking agents, we suggest that the synthesis of nE(2) is induced or enhanced by HFS through aromatase activation. In this context, the local production of nE(2) seems to be a very effective mechanism to modulate the amplitude of LTP.
Collapse
Affiliation(s)
- S Grassi
- Dipartimento di Medicina Interna, Sezione di Fisiologia Umana, Università di Perugia, Via del Giochetto, I-06126 Perugia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
106
|
Marin R. Signalosomes in the brain: relevance in the development of certain neuropathologies such as Alzheimer's disease. Front Physiol 2011; 2:23. [PMID: 21852974 PMCID: PMC3151622 DOI: 10.3389/fphys.2011.00023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 05/10/2011] [Indexed: 11/13/2022] Open
Abstract
Emerging data suggest that compartmentalization of signaling molecules into particular membrane compartments, or lipid rafts, may be at the basis of numerous activities related to neuronal preservation against different pathologies. These signaling platforms (signalosomes) are formed by complex lipid and protein that may interact to develop a plethora of different physiological responses upon activation by different extracellular stimuli, thereby contributing to neuroprotection. One of the first studied signalosomes involved in neuroprotection against Alzheimer's disease (AD) is constituted by estrogen receptor (ER), in association with scaffolding caveolin-1 and a voltage-dependent anion channel (VDAC). In this complex, ER plays a neuroprotective role partially through the modulation of VDAC activation, a porin involved in amyloid-beta-induced toxicity. Interestingly, ER and VDAC interactions appear to be altered in lipid rafts of AD brains, a phenomenon that may contribute to neuronal impairment. Alterations in lipid components of these subdomains may contribute to destabilization of this macrocomplex. These recent advances in the relevance of signaling platforms related to brain preservation, in particular against AD, are discussed in this work.
Collapse
Affiliation(s)
- Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Physiology, School of Medicine, La Laguna University Tenerife, Spain
| |
Collapse
|
107
|
Africander D, Verhoog N, Hapgood JP. Molecular mechanisms of steroid receptor-mediated actions by synthetic progestins used in HRT and contraception. Steroids 2011; 76:636-52. [PMID: 21414337 DOI: 10.1016/j.steroids.2011.03.001] [Citation(s) in RCA: 182] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 03/02/2011] [Accepted: 03/06/2011] [Indexed: 01/01/2023]
Abstract
Synthetic progestins are used by millions of women as contraceptives and in hormone replacement therapy (HRT), although their molecular mechanisms of action are not well understood. The importance of investigating these mechanisms, as compared to those of progesterone, has been highlighted by clinical evidence showing that medroxyprogesterone acetate (MPA), a first generation progestin, increases the risk of breast cancer and coronary heart disease in HRT users. A diverse range of later generation progestins with varying structures and pharmacological properties is available for therapeutic use and it is becoming clear that different progestins elicit beneficial and adverse effects to different extents. These differences in biological activity are likely to be due to many factors including variations in dose, metabolism, pharmacokinetics, bioavailability, and regulation of, and/or binding, to serum-binding proteins and steroidogenic enzymes. Since the intracellular effects on gene expression and cell signaling of steroids are mediated via intracellular steroid receptors, differential actions via the progesterone and other steroid receptors and their isoforms, are likely to be the major cause of differential intracellular actions of progestins. Since many progestins bind not only to the progesterone receptor, but also to the glucocorticoid, androgen, mineralocorticoid, and possibly the estrogen receptors, it is plausible that synthetic progestins exert therapeutic actions as well as side-effects via some of these receptors. Here we review the molecular mechanisms of intracellular actions of old (MPA, norethisterone, levonorgestrel, gestodene) vs. new (drospirenone, dienogest, trimegestone) generation progestins, via steroid receptors.
Collapse
Affiliation(s)
- Donita Africander
- Department of Biochemistry, University of Stellenbosch, Private Bag X1, Matieland, 7602, South Africa
| | | | | |
Collapse
|
108
|
Lim CKP, Ho CSH, Chou CHN, Waye MMY. Association of the rs3743205 variant of DYX1C1 with dyslexia in Chinese children. Behav Brain Funct 2011; 7:16. [PMID: 21599957 PMCID: PMC3123182 DOI: 10.1186/1744-9081-7-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 05/20/2011] [Indexed: 11/10/2022] Open
Abstract
Background Dyslexia is a learning disability that is characterized by difficulties in the acquisition of reading and spelling skills independent of intelligence, motivation or schooling. Studies of western populations have suggested that DYX1C1 is a candidate gene for dyslexia. In view of the different languages used in Caucasian and Chinese populations, it is therefore worthwhile to investigate whether there is an association of DYX1C1 in Chinese children with dyslexia. Method and Results Eight single nucleotide polymorphisms (SNPs) were genotyped from three hundred and ninety three individuals from 131 Chinese families with two which have been reported in the literature and six tag SNPs at DYX1C1. Analysis for allelic and haplotypic associations was performed with the UNPHASED program and multiple testing was corrected using false discovery rates. We replicated the previously reported association of rs3743205 in Chinese children with dyslexia (pcorrected = 0.0072). This SNP was also associated with rapid naming, phonological memory and orthographic skills in quantitative trait analysis. Conclusion Our findings suggest that DYX1C1 is associated with dyslexia in people of Chinese ethnicity in Hong Kong.
Collapse
Affiliation(s)
- Cadmon K P Lim
- Croucher Laboratory for Human Genomics, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong
| | | | | | | |
Collapse
|
109
|
Oestrogen: an overlooked mediator in the neuropsychopharmacology of treatment response? Int J Neuropsychopharmacol 2011; 14:553-66. [PMID: 20860875 DOI: 10.1017/s1461145710000982] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Major depression (MD) and anorexia nervosa (AN) often present comorbidly and both share some affective symptoms, despite obvious phenotypic differences. In the illness phase, pathophysiological evidence indicates similar abnormalities in both clinical groups including dysfunction in the serotonin (5-HT) system (of which some abnormalities persist following recovery) and between 60% and 80% of patients in both groups present with significant hyperactivity of the hypothalamo-pituitary-adrenal (HPA) axis. First-line approach to treatment for MD involves modulation of the 5-HT system using selective serotonin reuptake inhibitors (SSRIs). For AN, treatment with SSRIs has been shown to be considerably less effective compared to MD. Both illnesses show marked dysregulation in the HPA axis. A consequence of SSRI treatment is a reduction and/or normalization of indices of the HPA axis [i.e. cortisol, adrenocorticotropic hormone (ACTH)], which is consistent with recovery levels in both clinical groups. Oestrogen (in high doses) has been shown to exert antidepressant effects and positively impact on MD symptoms as a treatment in its own right, or in combination with antidepressants, in women of menopausal age. It is proposed that a combination of SSRIs and oestrogen therapy may facilitate physiological normalization in MD in women of non-menopausal age and in AN. Preliminary evidence suggests oestrogen treatment alone is of some benefit to patients and it is proposed that a combination of SSRI and oestrogen will precipitate and potentially accelerate symptomatic remission. Should this approach be successful, it offers the capacity for improvement over traditional antidepressant use in women diagnosed with MD and a novel strategy for the treatment of AN, a serious clinical illness associated with the highest mortality of any psychiatric condition.
Collapse
|
110
|
Paramanik V, Thakur MK. AIB1 shows variation in interaction with ERβTAD and expression as a function of age in mouse brain. Biogerontology 2011; 12:321-8. [PMID: 21442277 DOI: 10.1007/s10522-011-9330-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 03/12/2011] [Indexed: 11/29/2022]
Abstract
Estrogen mediates its multiple functions in the brain through the recruitment of a number of interacting proteins. In this paper, we report the identification of 160 kD interacting nuclear protein of estrogen receptor (ER)β-transactivation domain (TAD) as amplified in breast cancer 1(AIB1) by pull down assay, immunoblotting, far-western analysis and immunoprecipitation. Further we show the age dependent interaction and expression of AIB1 in the brain of young (6 weeks), adult (25 weeks) and old (70 weeks) AKR strain mice of both sexes. The immunoprecipitation data revealed higher interaction of AIB1 in young than adult and old male mice. In contrast, the interaction was low in young, increased in adult but decreased in old female. However, immunoblotting showed age related increase in the expression of AIB1 in both male and female mice. Further, the level of interaction of AIB1 with ERβTAD in young and old male was significantly higher than female of same age, whereas the expression of AIB1 in adult and old female was significantly higher than male of same age. These data suggest that such age dependent variation in the interaction of AIB1 with ERβTAD and its expression may be helpful to regulate estrogen-mediated gene functions during aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Vijay Paramanik
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Banaras Hindu University, Varanasi, 221 005, India
| | | |
Collapse
|
111
|
Yuen GS, McEwen BS, Akama KT. LIM kinase mediates estrogen action on the actin depolymerization factor Cofilin. Brain Res 2011; 1379:44-52. [PMID: 20696146 PMCID: PMC3021767 DOI: 10.1016/j.brainres.2010.07.067] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Revised: 07/15/2010] [Accepted: 07/18/2010] [Indexed: 12/24/2022]
Abstract
The ovarian hormone estrogen increases the axospinous synapse density in the hippocampal CA1 region of young female rats but fails to do so in aged rats. This estrogen-mediated alteration of spine synapse structures suggests the coincident requirement for the structural reorganization of the underlying actin cytoskeleton network. Actin reorganization is known to require the deactivation of Cofilin, an actin depolymerization factor. Cofilin is deactivated by LIM kinase (LIMK), and LIMK activity is modulated by the phosphorylation of specific residues. We have previously demonstrated that estrogen is able to increase phosphorylated LIMK (pLIMK) immunoreactivity (IR) in the hippocampus in vivo and that this estrogen-stimulated pLIMK-IR is decreased in the aged brain. Because Cofilin phosphorylation allows for actin filament elongation and spine synapse growth, we sought to determine if estrogen acts through Cofilin and if such estrogen action requires the observed LIMK activity. Using both hippocampal neurons and the NG108-15 neuroblastoma cell line, we demonstrate here that estrogen stimulates the phosphorylation of Cofilin in vitro. Furthermore, this estrogen action on Cofilin requires LIMK. Lastly, while initiating the phosphorylation of LIMK and Cofilin, estrogen can also stimulate the formation of filopodial extensions, an early step in the formation of nascent spines, demonstrating that estrogen can alter the actin-dependent neuronal morphology. This linkage of estrogen communication to Cofilin via LIMK provides the functionality to the age-sensitive pLIMK-IR that we have observed in vivo.
Collapse
Affiliation(s)
- Genevieve S Yuen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065-6399, USA
| | | | | |
Collapse
|
112
|
Kumar P, Taha A, Kale RK, Cowsik SM, Baquer NZ. Physiological and biochemical effects of 17β estradiol in aging female rat brain. Exp Gerontol 2011; 46:597-605. [PMID: 21377519 DOI: 10.1016/j.exger.2011.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2010] [Revised: 01/13/2011] [Accepted: 02/22/2011] [Indexed: 01/10/2023]
Abstract
Aging in females and males is considered as the end of natural protection against age related diseases like osteoporosis, coronary heart disease, diabetes, Alzheimer's disease and Parkinson's disease. These changes increase during menopausal condition in females when the level of estradiol is decreased. The objective of this study was to observe the changes in activities of monoamine oxidase, glucose transporter-4 levels, membrane fluidity, lipid peroxidation levels and lipofuscin accumulation occurring in brains of female rats of 3 months (young), 12 months (adult) and 24 months (old) age groups, and to see whether these changes are restored to normal levels after exogenous administration of estradiol (0.1 μg/g body weight for 1 month). The results obtained in the present work revealed that normal aging was associated with significant increases in the activity of monoamine oxidase, lipid peroxidation levels and lipofuscin accumulation in the brains of aging female rats, and a decrease in glucose transporter-4 level and membrane fluidity. Our data showed that estradiol treatment significantly decreased monoamine oxidase activity, lipid peroxidation and lipofuscin accumulation in brain regions of aging rats, and a reversal of glucose transporter-4 levels and membrane fluidity was achieved, therefore it can be concluded from the present findings that estradiol's beneficial effects seemed to arise from its antilipofuscin, antioxidant and antilipidperoxidative effects, implying an overall anti-aging action. The results of this study will be useful for pharmacological modification of the aging process and applying new strategies for control of age related disorders.
Collapse
Affiliation(s)
- Pardeep Kumar
- School of Life Sciences, Jawaharlal Nehru University, 110067, New Delhi, India
| | | | | | | | | |
Collapse
|
113
|
Chen L, Mei XP, Gao CJ, Zhang GH, Sun XD. Histologic distribution, fragment cloning, and sequence analysis of g protein couple receptor 30 in rat submaxillary gland. Anat Rec (Hoboken) 2011; 294:706-11. [PMID: 21365775 DOI: 10.1002/ar.21349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 12/14/2010] [Accepted: 12/15/2010] [Indexed: 11/07/2022]
Abstract
Recent studies indicated that G protein couple receptor 30 (GPR30), a nongenomic estrogen receptor, is widely expressed in many organ systems inducing many quick reaction of estrogen. However, there was rare report about the expression of GPR30 in the salivary gland. In the present study, we investigated the distribution of GPR30 in rat submaxillary gland by means of immunohistochemistry and in situ hybridization. GPR30 core sequences were amplified by RT-PCR with total RNA extracted from rat submaxillary gland and were analyzed by sequencing with Sanger's method. The results showed that the epithelial cells of serous alveoli and granular convoluted duct in rat submaxillary gland displayed GPR30-immunoreactivity on the plasma membrane and cytoplasm. Moreover, GPR30 mRNA hybridization signals were also detected in the cytoplasm of the above cells. GPR30 cDNA sequence cloned from rat submaxillary gland is identical to that of GPR30 from rat paraventricular and supraoptic nucleus. In conclusion, the expression of GPR30 in the serous and granular epithelial cells of submaxillary gland indicates that submaxillary gland could also be a target organ rapidly responding to estrogen stimulus, and estrogen may be involved in the functional regulation of submaxillary gland.
Collapse
Affiliation(s)
- Lei Chen
- Department of Gynaecology and Obstetrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | |
Collapse
|
114
|
Jeng YJ, Watson CS. Combinations of physiologic estrogens with xenoestrogens alter ERK phosphorylation profiles in rat pituitary cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2011; 119:104-12. [PMID: 20870566 PMCID: PMC3018487 DOI: 10.1289/ehp.1002512] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 09/22/2010] [Indexed: 05/08/2023]
Abstract
BACKGROUND Estrogens are potent nongenomic phospho-activators of extracellular-signal-regulated kinases (ERKs). A major concern about the toxicity of xenoestrogens (XEs) is potential alteration of responses to physiologic estrogens when XEs are present simultaneously. OBJECTIVES We examined estrogen-induced ERK activation, comparing the abilities of structurally related XEs (alkylphenols and bisphenol A) to alter ERK responses induced by physiologic concentrations (1 nM) of estradiol (E2), estrone (E1), and estriol (E3). METHODS We quantified hormone/mimetic-induced ERK phosphorylations in the GH3/B6/F10 rat pituitary cell line using a plate immunoassay, comparing effects with those on cell proliferation and by estrogen receptor subtype-selective ligands. RESULTS Alone, these structurally related XEs activate ERKs in an oscillating temporal pattern similar (but not identical) to that with physiologic estrogens. The potency of all estrogens was similar (active between femtomolar and nanomolar concentrations). XEs potently disrupted physiologic estrogen signaling at low, environmentally relevant concentrations. Generally, XEs potentiated (at the lowest, subpicomolar concentrations) and attenuated (at the highest, picomolar to 100 nM concentrations) the actions of the physiologic estrogens. Some XEs showed pronounced nonmonotonic responses/inhibitions. The phosphorylated ERK and proliferative responses to receptor-selective ligands were only partially correlated. CONCLUSIONS XEs are both imperfect potent estrogens and endocrine disruptors; the more efficacious an XE, the more it disrupts actions of physiologic estrogens. This ability to disrupt physiologic estrogen signaling suggests that XEs may disturb normal functioning at life stages where actions of particular estrogens are important (e.g., development, reproductive cycling, pregnancy, menopause).
Collapse
Affiliation(s)
| | - Cheryl S. Watson
- Address correspondence to C.S. Watson, Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0645 USA. Telephone/fax: (409) 772-2383. E-mail:
| |
Collapse
|
115
|
Srivastava DP, Penzes P. Rapid estradiol modulation of neuronal connectivity and its implications for disease. Front Endocrinol (Lausanne) 2011; 2:77. [PMID: 22654827 PMCID: PMC3356153 DOI: 10.3389/fendo.2011.00077] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 11/04/2011] [Indexed: 12/05/2022] Open
Abstract
Estrogens have multiple actions in the brain including modulating synaptic plasticity, connectivity, and cognitive behaviors. While the classical view of estrogens are as endocrine signals, whose effects manifest via the regulation of gene transcription, mounting evidence has been presented demonstrating that estrogens have rapid effects within specific areas of the brain. The emergence that 17 β-estradiol can be produced locally in the brain which can elicit rapid (within minutes) cellular responses has led to its classification as a neurosteroid. Moreover, recent studies have also begun to detail the molecular and cellular underpinnings of how 17 β-estradiol can rapidly modulate spiny synapses (dendritic spines). Remodeling of dendritic spines is a key step in the rewiring of neuronal circuitry thought to underlie the processing and storage of information in the forebrain. Conversely, abnormal remodeling of dendritic spines is thought to contribute to a number of psychiatric and neurodevelopmental disorders. Here we review recent molecular and cellular work that offers a potential mechanism of how 17 β-estradiol may modulate synapse structure and function of cortical neurons. This mechanism allows cortical neurons to respond to activity-dependent stimuli with greater efficacy. In turn this form of plasticity may provide an insight into how 17 β-estradiol can modulate the rewiring of neuronal circuits, underlying its ability to influencing cortically based behaviors. We will then go on to discuss the potential role of 17 β-estradiol modulation of neural circuits and its potential relevance for the treatment of psychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Deepak P. Srivastava
- Department of Physiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
- Department of Neuroscience and Centre for the Cellular Basis of Behaviour, The James Black Centre, Institute of Psychiatry, King’s College LondonLondon, UK
- *Correspondence: Deepak P. Srivastava, Department of Physiology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, USA. e-mail:
| | - Peter Penzes
- Department of Physiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| |
Collapse
|
116
|
Bourque M, Dluzen DE, Di Paolo T. Male/Female differences in neuroprotection and neuromodulation of brain dopamine. Front Endocrinol (Lausanne) 2011; 2:35. [PMID: 22654803 PMCID: PMC3356083 DOI: 10.3389/fendo.2011.00035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 09/02/2011] [Indexed: 12/26/2022] Open
Abstract
The existence of a sex difference in Parkinson's disease (PD) is observed as related to several variables, including susceptibility of the disease, age at onset, and symptoms. These differences between men and women represent a significant characteristic of PD, which suggest that estrogens may exert beneficial effects against the development and the progression of the disease. This paper reviews the neuroprotective and neuromodulator effects of 17β-estradiol and progesterone as compared to androgens in the nigrostriatal dopaminergic (NSDA) system of both female and male rodents. The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mice model of PD and methamphetamine toxicity faithfully reproduce the sex differences of PD in that endogenous estrogen levels appear to influence the vulnerability to toxins targeting the NSDA system. Exogenous 17β-estradiol and/or progesterone treatments show neuroprotective properties against NSDA toxins while androgens fail to induce any beneficial effect. Sex steroid treatments show male and female differences in their neuroprotective action against methamphetamine toxicity. NSDA structure and function, as well as the distribution of estrogen receptors, show sex differences and may influence the susceptibility to the toxins and the response to sex steroids. Genomic and non-genomic actions of 17β-estradiol converge to promote survival factors and the presence of both estrogen receptors α and β are critical to 17β-estradiol neuroprotective action against MPTP toxicity.
Collapse
Affiliation(s)
- Mélanie Bourque
- Molecular Endocrinology and Genomic Research Center, Centre de recherche du CHUQ (CHUL)Quebec City, QC, Canada
- Faculty of Pharmacy, Laval University, Quebec CityQC, Canada
| | - Dean E. Dluzen
- Department of Anatomy and Neurobiology, Northeastern Ohio Universities College of Medicine and PharmacyRootstown, OH, USA
| | - Thérèse Di Paolo
- Molecular Endocrinology and Genomic Research Center, Centre de recherche du CHUQ (CHUL)Quebec City, QC, Canada
- Faculty of Pharmacy, Laval University, Quebec CityQC, Canada
- *Correspondence: Thérèse Di Paolo, Molecular Endocrinology and Genomic Research Center, Centre de recherche du CHUQ (CHUL), 2705 Laurier Boulevard, Quebec City, QC, Canada G1V 4G2. e-mail:
| |
Collapse
|
117
|
Identification of 9-cis-retinoic acid as a pancreas-specific autacoid that attenuates glucose-stimulated insulin secretion. Proc Natl Acad Sci U S A 2010; 107:21884-9. [PMID: 21115832 DOI: 10.1073/pnas.1008859107] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The all-trans-retinoic acid (atRA) isomer, 9-cis-retinoic acid (9cRA), activates retinoic acid receptors (RARs) and retinoid X receptors (RXRs) in vitro. RARs control multiple genes, whereas RXRs serve as partners for RARs and other nuclear receptors that regulate metabolism. Physiological function has not been determined for 9cRA, because it has not been detected in serum or multiple tissues with analytically validated assays. Here, we identify 9cRA in mouse pancreas by liquid chromatography/tandem mass spectrometry (LC/MS/MS), and show that 9cRA decreases with feeding and after glucose dosing and varies inversely with serum insulin. 9cRA reduces glucose-stimulated insulin secretion (GSIS) in mouse islets and in the rat β-cell line 832/13 within 15 min by reducing glucose transporter type 2 (Glut2) and glucokinase (GK) activities. 9cRA also reduces Pdx-1 and HNF4α mRNA expression, ∼8- and 80-fold, respectively: defects in Pdx-1 or HNF4α cause maturity onset diabetes of the young (MODY4 and 1, respectively), as does a defective GK gene (MODY2). Pancreas β-cells generate 9cRA, and mouse models of reduced β-cell number, heterozygous Akita mice, and streptozotocin-treated mice have reduced 9cRA. 9cRA is abnormally high in glucose-intolerant mice, which have β-cell hypertropy, including mice with diet-induced obesity (DIO) and ob/ob and db/db mice. These data establish 9cRA as a pancreas-specific autacoid with multiple mechanisms of action and provide unique insight into GSIS.
Collapse
|
118
|
Siriphorn A, Chompoopong S, Floyd CL. 17β-estradiol protects Schwann cells against H2O2-induced cytotoxicity and increases transplanted Schwann cell survival in a cervical hemicontusion spinal cord injury model. J Neurochem 2010; 115:864-72. [PMID: 20456002 DOI: 10.1111/j.1471-4159.2010.06770.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Schwann cell (SC) transplantation is a promising repair strategy after spinal cord injury (SCI); however, a large number of SCs do not survive following transplantation. Previous studies have shown that 17β-estradiol (E2) protects several cell types against cytotoxicity. Thus, this study evaluated the protective potential of E2 on SCs in vitro and investigated the effect of E2 on transplanted SC survival in a rat model of SCI. Primary SC cultures were found to robustly express estrogen receptors (ER) and incubation with E2 protected SCs against hydrogen peroxide-induced cell death. This protection was not inhibited by the ER antagonist ICI 182,780, suggesting that genomic signaling is not necessary for protection. In a subsequent experiment, cervical hemicontusion SCI was induced in male rats followed by sustained administration of E2 or placebo. Eight days after SCI, SCs were transplanted into the injury epicenter. E2 treatment significantly increased the number of surviving labeled transplanted SCs evaluated 7 days after transplantation. These data demonstrate that E2 protects SCs against oxidative stress and improves transplanted SC survival, which suggests that E2 administration may be an intervention of choice for enhancing survival of transplanted SCs after SCI.
Collapse
Affiliation(s)
- Akkradate Siriphorn
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | |
Collapse
|
119
|
Influence of different estrogens on neuroplasticity and cognition in the hippocampus. Biochim Biophys Acta Gen Subj 2010; 1800:1056-67. [DOI: 10.1016/j.bbagen.2010.01.006] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 01/13/2010] [Accepted: 01/16/2010] [Indexed: 11/18/2022]
|
120
|
Henderson VW. Action of estrogens in the aging brain: Dementia and cognitive aging. Biochim Biophys Acta Gen Subj 2010; 1800:1077-83. [DOI: 10.1016/j.bbagen.2009.11.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2009] [Revised: 10/20/2009] [Accepted: 11/02/2009] [Indexed: 10/20/2022]
|
121
|
Abstract
The ATP-driven efflux transporter, breast cancer resistance protein (BCRP), handles many therapeutic drugs, including chemotherapeutics, limiting their ability to cross the blood-brain barrier. This study provides new insight into rapid, nongenomic regulation of BCRP transport activity at the blood-brain barrier. Using isolated brain capillaries from rats and mice as an ex vivo blood-brain barrier model, we show that BCRP protein is highly expressed in brain capillary membranes and functionally active in intact capillaries. We show that nanomolar concentrations of 17-β-estradiol (E2) rapidly reduced BCRP transport activity in the brain capillaries. This E2-mediated effect occurred within minutes and did not involve transcription, translation, or proteasomal degradation, indicating a nongenomic mechanism. Removing E2 after 1 h fully reversed the loss of BCRP activity. Experiments using agonists and antagonists for estrogen receptor (ER)α and ERβ and brain capillaries from ERα and ERβ knockout mice demonstrated that E2 could signal through either receptor to reduce BCRP transport function. We speculate that this nongenomic E2-signaling pathway could potentially be used for targeting BCRP at the blood-brain barrier, in brain tumors, and in brain tumor stem cells to improve chemotherapy of the central nervous system.
Collapse
|
122
|
Arterburn JB, Oprea TI, Prossnitz ER, Edwards BS, Sklar LA. Discovery of selective probes and antagonists for G-protein-coupled receptors FPR/FPRL1 and GPR30. Curr Top Med Chem 2010; 9:1227-36. [PMID: 19807662 DOI: 10.2174/156802609789753608] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 07/29/2009] [Indexed: 01/04/2023]
Abstract
Recent technological advances in flow cytometry provide a versatile platform for high throughput screening of compound libraries coupled with high-content biological testing and drug discovery. The G protein-coupled receptors (GPCRs) constitute the largest class of signaling molecules in the human genome with frequent roles in disease pathogenesis, yet many examples of orphan receptors with unknown ligands remain. The complex biology and potential for drug discovery within this class provide strong incentives for chemical biology approaches seeking to develop small molecule probes to facilitate elucidation of mechanistic pathways and enable specific manipulation of the activity of individual receptors. We have initiated small molecule probe development projects targeting two distinct families of GPCRs: the formylpeptide receptors (FPR/FPRL1) and G protein-coupled estrogen receptor (GPR30). In each case the assay for compound screening involved the development of an appropriate small molecule fluorescent probe, and the flow cytometry platform provided inherently biological rich assays that enhanced the process of identification and optimization of novel antagonists. The contributions of cheminformatics analysis tools, virtual screening, and synthetic chemistry in synergy with the biomolecular screening program have yielded valuable new chemical probes with high binding affinity, selectivity for the targeted receptor, and potent antagonist activity. This review describes the discovery of novel small molecule antagonists of FPR and FPRL1, and GPR30, and the associated characterization process involving secondary assays, cell based and in vivo studies to define the selectivity and activity of the resulting chemical probes.
Collapse
Affiliation(s)
- Jeffrey B Arterburn
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM 88003, USA.
| | | | | | | | | |
Collapse
|
123
|
Estrogen receptor beta-selective agonists stimulate calcium oscillations in human and mouse embryonic stem cell-derived neurons. PLoS One 2010; 5:e11791. [PMID: 20668547 PMCID: PMC2910705 DOI: 10.1371/journal.pone.0011791] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Accepted: 06/18/2010] [Indexed: 12/30/2022] Open
Abstract
Estrogens are used extensively to treat hot flashes in menopausal women. Some of the beneficial effects of estrogens in hormone therapy on the brain might be due to nongenomic effects in neurons such as the rapid stimulation of calcium oscillations. Most studies have examined the nongenomic effects of estrogen receptors (ER) in primary neurons or brain slices from the rodent brain. However, these cells can not be maintained continuously in culture because neurons are post-mitotic. Neurons derived from embryonic stem cells could be a potential continuous, cell-based model to study nongenomic actions of estrogens in neurons if they are responsive to estrogens after differentiation. In this study ER-subtype specific estrogens were used to examine the role of ERα and ERβ on calcium oscillations in neurons derived from human (hES) and mouse embryonic stem cells. Unlike the undifferentiated hES cells the differentiated cells expressed neuronal markers, ERβ, but not ERα. The non-selective ER agonist 17β-estradiol (E2) rapidly increased [Ca2+]i oscillations and synchronizations within a few minutes. No change in calcium oscillations was observed with the selective ERα agonist 4,4′,4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT). In contrast, the selective ERβ agonists, 2,3-bis(4-Hydroxyphenyl)-propionitrile (DPN), MF101, and 2-(3-fluoro-4-hydroxyphenyl)-7-vinyl-1,3 benzoxazol-5-ol (ERB-041; WAY-202041) stimulated calcium oscillations similar to E2. The ERβ agonists also increased calcium oscillations and phosphorylated PKC, AKT and ERK1/2 in neurons derived from mouse ES cells, which was inhibited by nifedipine demonstrating that ERβ activates L-type voltage gated calcium channels to regulate neuronal activity. Our results demonstrate that ERβ signaling regulates nongenomic pathways in neurons derived from ES cells, and suggest that these cells might be useful to study the nongenomic mechanisms of estrogenic compounds.
Collapse
|
124
|
Iwakura T, Iwafuchi M, Muraoka D, Yokosuka M, Shiga T, Watanabe C, Ohtani-Kaneko R. In vitro effects of bisphenol A on developing hypothalamic neurons. Toxicology 2010; 272:52-8. [DOI: 10.1016/j.tox.2010.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 04/03/2010] [Accepted: 04/09/2010] [Indexed: 12/29/2022]
|
125
|
Gillies GE, McArthur S. Estrogen actions in the brain and the basis for differential action in men and women: a case for sex-specific medicines. Pharmacol Rev 2010; 62:155-98. [PMID: 20392807 PMCID: PMC2879914 DOI: 10.1124/pr.109.002071] [Citation(s) in RCA: 502] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The classic view of estrogen actions in the brain was confined to regulation of ovulation and reproductive behavior in the female of all mammalian species studied, including humans. Burgeoning evidence now documents profound effects of estrogens on learning, memory, and mood as well as neurodevelopmental and neurodegenerative processes. Most data derive from studies in females, but there is mounting recognition that estrogens play important roles in the male brain, where they can be generated from circulating testosterone by local aromatase enzymes or synthesized de novo by neurons and glia. Estrogen-based therapy therefore holds considerable promise for brain disorders that affect both men and women. However, as investigations are beginning to consider the role of estrogens in the male brain more carefully, it emerges that they have different, even opposite, effects as well as similar effects in male and female brains. This review focuses on these differences, including sex dimorphisms in the ability of estradiol to influence synaptic plasticity, neurotransmission, neurodegeneration, and cognition, which, we argue, are due in a large part to sex differences in the organization of the underlying circuitry. There are notable sex differences in the incidence and manifestations of virtually all central nervous system disorders, including neurodegenerative disease (Parkinson's and Alzheimer's), drug abuse, anxiety, and depression. Understanding the cellular and molecular basis of sex differences in brain physiology and responses to estrogen and estrogen mimics is, therefore, vitally important for understanding the nature and origins of sex-specific pathological conditions and for designing novel hormone-based therapeutic agents that will have optimal effectiveness in men or women.
Collapse
Affiliation(s)
- Glenda E Gillies
- Centre for Neuroscience, Department of Medicine, Hammersmith Hospital, Imperial College Faculty of Medicine, DuCane Road, London W12ONN, UK.
| | | |
Collapse
|
126
|
Numakawa T, Yokomaku D, Richards M, Hori H, Adachi N, Kunugi H. Functional interactions between steroid hormones and neurotrophin BDNF. World J Biol Chem 2010; 1:133-43. [PMID: 21540998 PMCID: PMC3083963 DOI: 10.4331/wjbc.v1.i5.133] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 05/20/2010] [Accepted: 05/24/2010] [Indexed: 02/05/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a critical neurotrophin, regulates many neuronal aspects including cell differentiation, cell survival, neurotransmission, and synaptic plasticity in the central nervous system (CNS). Though BDNF has two types of receptors, high affinity tropomyosin-related kinase (Trk)B and low affinity p75 receptors, BDNF positively exerts its biological effects on neurons via activation of TrkB and of resultant intracellular signaling cascades including mitogen-activated protein kinase/extracellular signal-regulated protein kinase, phospholipase Cγ, and phosphoinositide 3-kinase pathways. Notably, it is possible that alteration in the expression and/or function of BDNF in the CNS is involved in the pathophysiology of various brain diseases such as stroke, Parkinson’s disease, Alzheimer’s disease, and mental disorders. On the other hand, glucocorticoids, stress-induced steroid hormones, also putatively contribute to the pathophysiology of depression. Interestingly, in addition to the reduction in BDNF levels due to increased glucocorticoid exposure, current reports demonstrate possible interactions between glucocorticoids and BDNF-mediated neuronal functions. Other steroid hormones, such as estrogen, are involved in not only sexual differentiation in the brain, but also numerous neuronal events including cell survival and synaptic plasticity. Furthermore, it is well known that estrogen plays a role in the pathophysiology of Parkinson’s disease, Alzheimer’s disease, and mental illness, while serving to regulate BDNF expression and/or function. Here, we present a broad overview of the current knowledge concerning the association between BDNF expression/function and steroid hormones (glucocorticoids and estrogen).
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Tadahiro Numakawa, Misty Richards, Hiroaki Hori, Naoki Adachi, Hiroshi Kunugi, Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | | | | | | | | | | |
Collapse
|
127
|
Yang LC, Zhang QG, Zhou CF, Yang F, Zhang YD, Wang RM, Brann DW. Extranuclear estrogen receptors mediate the neuroprotective effects of estrogen in the rat hippocampus. PLoS One 2010; 5:e9851. [PMID: 20479872 PMCID: PMC2866326 DOI: 10.1371/journal.pone.0009851] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 03/04/2010] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND 17beta-estradiol (E2) has been implicated to exert neuroprotective effects in the brain following cerebral ischemia. Classically, E2 is thought to exert its effects via genomic signaling mediated by interaction with nuclear estrogen receptors. However, the role and contribution of extranuclear estrogen receptors (ER) is unclear and was the subject of the current study. METHODOLOGY/PRINCIPAL FINDINGS To accomplish this goal, we employed two E2 conjugates (E2 dendrimer, EDC, and E2-BSA) that can interact with extranuclear ER and exert rapid nongenomic signaling, but lack the ability to interact with nuclear ER due to their inability to enter the nucleus. EDC or E2-BSA (10 microM) was injected icv 60 min prior to global cerebral ischemia (GCI). FITC-tagged EDC or E2-BSA revealed high uptake in the hippocampal CA1 region after icv injection, with a membrane (extranuclear) localization pattern in cells. Both EDC and E2-BSA exerted robust neuroprotection in the CA1 against GCI, and the effect was blocked by the ER antagonist, ICI182,780. EDC and E2-BSA both rapidly enhanced activation of the prosurvival kinases, ERK and Akt, while attenuating activation of the proapoptotic kinase, JNK following GCI, effects that were blocked by ICI182,780. Administration of an MEK or PI3K inhibitor blocked the neuroprotective effects of EDC and E2-BSA. Further studies showed that EDC increased p-CREB and BDNF in the CA1 region in an ERK- and Akt-dependent manner, and that cognitive outcome after GCI was preserved by EDC in an ER-dependent manner. CONCLUSIONS/SIGNIFICANCE In conclusion, the current study demonstrates that activation of extranuclear ER results in induction of ERK-Akt-CREB-BDNF signaling in the hippocampal CA1 region, which significantly reduces ischemic neuronal injury and preserves cognitive function following GCI. The study adds to a growing literature that suggests that extranuclear ER can have important actions in the brain.
Collapse
Affiliation(s)
- Li-cai Yang
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Quan-Guang Zhang
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Cai-feng Zhou
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Fang Yang
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Yi-dong Zhang
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Rui-min Wang
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Darrell W. Brann
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia, United States of America
| |
Collapse
|
128
|
Burai R, Ramesh C, Shorty M, Curpan R, Bologa C, Sklar LA, Oprea T, Prossnitz ER, Arterburn JB. Highly efficient synthesis and characterization of the GPR30-selective agonist G-1 and related tetrahydroquinoline analogs. Org Biomol Chem 2010; 8:2252-9. [PMID: 20401403 PMCID: PMC2913306 DOI: 10.1039/c001307b] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The GPR30 agonist probe G-1 and structural analogs were efficiently synthesized using multicomponent or stepwise Sc(III)-catalyzed aza-Diels-Alder cyclization. Optimization of solvent and reaction temperature provided enhanced endo-diastereoselectivity.
Collapse
Affiliation(s)
- Ritwik Burai
- Department of Chemistry and Biochemistry MSC 3C, New Mexico State University, P.O. Box 30001, Las Cruces, New Mexico 88003
| | - Chinnasamy Ramesh
- Department of Chemistry and Biochemistry MSC 3C, New Mexico State University, P.O. Box 30001, Las Cruces, New Mexico 88003
| | - Marvin Shorty
- Department of Chemistry and Biochemistry MSC 3C, New Mexico State University, P.O. Box 30001, Las Cruces, New Mexico 88003
| | - Ramona Curpan
- Institute of Chemistry, Romanian Academy, Timisoara, 300223 Romania
- Division of Biocomputing, Department of Biochemistry & Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Cristian Bologa
- Division of Biocomputing, Department of Biochemistry & Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Larry A. Sklar
- Cancer Research and Treatment Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Tudor Oprea
- Division of Biocomputing, Department of Biochemistry & Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- Cancer Research and Treatment Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Eric R. Prossnitz
- Cancer Research and Treatment Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- Department of Cell Biology & Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Jeffrey B. Arterburn
- Department of Chemistry and Biochemistry MSC 3C, New Mexico State University, P.O. Box 30001, Las Cruces, New Mexico 88003
- Cancer Research and Treatment Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| |
Collapse
|
129
|
Bredfeldt TG, Greathouse KL, Safe SH, Hung MC, Bedford MT, Walker CL. Xenoestrogen-induced regulation of EZH2 and histone methylation via estrogen receptor signaling to PI3K/AKT. Mol Endocrinol 2010; 24:993-1006. [PMID: 20351197 PMCID: PMC2870935 DOI: 10.1210/me.2009-0438] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 02/18/2010] [Indexed: 12/17/2022] Open
Abstract
Although rapid, membrane-activated estrogen receptor (ER) signaling is no longer controversial, the biological function of this nongenomic signaling is not fully characterized. We found that rapid signaling from membrane-associated ER regulates the histone methyltransferase enhancer of Zeste homolog 2 (EZH2). In response to both 17beta-estradiol (E2) and the xenoestrogen diethylstilbestrol, ER signaling via phosphatidylinositol 3-kinase/protein kinase B phosphorylates EZH2 at S21, reducing levels of trimethylation of lysine 27 on histone H3 in hormone-responsive cells. During windows of uterine development that are susceptible to developmental reprogramming, activation of this ER signaling pathway by diethylstilbestrol resulted in phosphorylation of EZH2 and reduced levels of trimethylation of lysine 27 on histone H3 in chromatin of the developing uterus. Furthermore, activation of nongenomic signaling reprogrammed the expression profile of estrogen-responsive genes in uterine myometrial cells, suggesting this as a potential mechanism for developmental reprogramming caused by early-life exposure to xenoestrogens. These data demonstrate that rapid ER signaling provides a direct linkage between xenoestrogen-induced nuclear hormone receptor signaling and modulation of the epigenetic machinery during tissue development.
Collapse
Affiliation(s)
- Tiffany G Bredfeldt
- Department of Carcinogenesis, University of Texas, M.D. Anderson Cancer Center, Science Park Research Division, 1808 Park Road 1C, P.O. Box 389, Smithville, Texas 78957, USA
| | | | | | | | | | | |
Collapse
|
130
|
Kumar S, Lata K, Mukhopadhyay S, Mukherjee TK. Role of estrogen receptors in pro-oxidative and anti-oxidative actions of estrogens: a perspective. Biochim Biophys Acta Gen Subj 2010; 1800:1127-35. [PMID: 20434525 DOI: 10.1016/j.bbagen.2010.04.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 03/23/2010] [Accepted: 04/21/2010] [Indexed: 01/07/2023]
Abstract
BACKGROUND Estrogens are steroid hormones responsible for the primary and secondary sexual characteristics in females. While pre-menopausal women use estrogens as the main constituents of contraceptive pills, post-menopausal women use the same for Hormone Replacement Therapy. Estrogens produce reactive oxygen species by increasing mitochondrial activity and redox cycling of estrogen metabolites. The phenolic hydroxyl group present at the C3 position of the A ring of estrogens can get oxidized either by accepting an electron or by losing a proton. Thus, estrogens might act as pro-oxidant in some settings, resulting in complicated non-communicable diseases, namely, cancer and cardiovascular disorders. However, in some other settings the phenolic hydroxyl group of estrogens may be responsible for the anti-oxidative beneficial functions and thus protect against cardiovascular and neurodegenerative diseases. SCOPE OF REVIEW To date, no single review article has mentioned the implication of estrogen receptors in both the pro-oxidative and anti-oxidative actions of estrogens. MAJOR CONCLUSION The controversial role of estrogens as pro-oxidant or anti-oxidant is largely dependent on cell types, ratio of different types of estrogen receptors present in a particular cell and context specificity of the estrogen hormone responses. Both pro-oxidant and anti-oxidant effects of estrogens might involve different estrogen receptors that can have either genomic or non-genomic action to manifest further hormonal response. GENERAL SIGNIFICANCE This review highlights the role of estrogen receptors in the pro-oxidative and anti-oxidative actions of estrogens with special emphasis on neuronal cells.
Collapse
Affiliation(s)
- Sukhdeep Kumar
- Department of Biology, Indiana Institute of Science Eduaction and Research (IISER), Transit Campus, Chandigarh, India
| | | | | | | |
Collapse
|
131
|
Zhang B, Subramanian S, Dziennis S, Jia J, Uchida M, Akiyoshi K, Migliati E, Lewis AD, Vandenbark AA, Offner H, Hurn PD. Estradiol and G1 reduce infarct size and improve immunosuppression after experimental stroke. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:4087-94. [PMID: 20304826 PMCID: PMC3142781 DOI: 10.4049/jimmunol.0902339] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Reduced risk and severity of stroke in adult females is thought to depend on normal endogenous levels of estrogen, a well-known neuroprotectant and immunomodulator. In male mice, experimental stroke induces immunosuppression of the peripheral immune system, characterized by a reduction in spleen size and cell numbers and decreased cytokine and chemokine expression. However, stroke-induced immunosuppression has not been evaluated in female mice. To test the hypothesis that estradiol (E2) deficiency exacerbates immunosuppression after focal stroke in females, we evaluated the effect of middle cerebral artery occlusion on infarct size and peripheral and CNS immune responses in ovariectomized mice with or without sustained, controlled levels of 17-beta-E2 administered by s.c. implant or the putative membrane estrogen receptor agonist, G1. Both E2- and G1-replacement decreased infarct volume and partially restored splenocyte numbers. Moreover, E2-replacement increased splenocyte proliferation in response to stimulation with anti-CD3/CD28 Abs and normalized aberrant mRNA expression for cytokines, chemokines, and chemokine receptors and percentage of CD4(+)CD25(+)FoxP3(+) T regulatory cells observed in E2-deficient animals. These beneficial changes in peripheral immunity after E2 replacement were accompanied by a profound reduction in expression of the chemokine, MIP-2, and a 40-fold increased expression of CCR7 in the lesioned brain hemisphere. These results demonstrate for the first time that E2 replacement in ovariectomized female mice improves stroke-induced peripheral immunosuppression.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Anesthesiology and Peri-Operative Medicine, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Wong CO, Huang Y, Yao X. Genistein potentiates activity of the cation channel TRPC5 independently of tyrosine kinases. Br J Pharmacol 2010; 159:1486-96. [PMID: 20233211 DOI: 10.1111/j.1476-5381.2010.00636.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE TRPC5 is a Ca(2+)-permeable channel with multiple modes of activation. We have explored the effects of genistein, a plant-derived isoflavone, on TRPC5 activity, and the mechanism(s) involved. EXPERIMENTAL APPROACH Effects of genistein on TRPC5 channels were investigated in TRPC5-over-expressing human embryonic kidney 293 (HEK) cells and bovine aortic endothelial cells (BAECs) using fluorescent Ca(2+) imaging and electrophysiological techniques. KEY RESULTS In TRPC5-over-expressing HEK cells, genistein stimulated TRPC5-mediated Ca(2+) influx, concentration dependently (EC(50)= 93 microM). Genistein and lanthanum activated TRPC5 channels synergistically. Effects of genistein on TRPC5 channels were mimicked by daidzein (100 microM), a genistein analogue inactive as a tyrosine kinase inhibitor, but not by known tyrosine kinase inhibitors herbimycin (2 microM), PP2 (20 microM) and lavendustin A (10 microM). Action of genistein on TRPC5 channels was not affected by an oestrogen receptor inhibitor ICI-182780 (50 microM) or a phospholipase C inhibitor U73122 (10 microM), suggesting genistein did not act through oestrogen receptors or phospholipase C. In BAECs, genistein (100 microM) stimulated TRPC5-mediated Ca(2+) influx. In patch clamp studies, both genistein (50 microM) and daidzein (50 microM) augmented TRPC5-mediated whole-cell cation current in TRPC5 over-expressing HEK cells. Genistein stimulated TRPC5 channel activity in excised inside-out membrane patch, suggesting that its action was relatively direct and did not require cytosolic factors. CONCLUSIONS AND IMPLICATIONS The present study is the first to demonstrate stimulation of a TRP channel by isoflavones. Genistein is a lipophilic compound able to stimulate TRPC5 activity in TRPC5-over-expressing HEK cells and in native vascular endothelial cells.
Collapse
Affiliation(s)
- Ching-On Wong
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | | | | |
Collapse
|
133
|
Estrogen is renoprotective via a nonreceptor-dependent mechanism after cardiac arrest in vivo. Anesthesiology 2010; 112:395-405. [PMID: 20068453 DOI: 10.1097/aln.0b013e3181c98da9] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Severe ischemia induces renal injury less frequently in women than men. In this study, cardiac arrest and cardiopulmonary resuscitation were used to assess whether estradiol is renoprotective via an estrogen receptor (ER)-dependent mechanism. MATERIALS AND METHODS Male and female C57BL/6 and ER gene-deleted mice underwent 10 min of cardiac arrest followed by cardiopulmonary resuscitation. Serum chemistries and renal stereology were measured 24 h after arrest. RESULTS Estrogen did not affect mean arterial pressure, regional renal cortical blood flow, and arterial blood gases. Hence, female kidneys were protected (mean +/- SEM: blood urea nitrogen, 65+/- 21 vs.149+/- 27 mg/dl, P = 0.04; creatinine, 0.14 +/- 0.05 vs. 0.73 +/- 0.16 mg/dl, P = 0.01; volume of necrotic tubules, 7 +/- 1% vs. 10 +/- 0%, P = 0.04). Estrogen also reduced renal injury. In intact females (n = 5), ovariectomized/vehicle-treated (n = 8), and ovariectomized/estrogen-treated (n = 8) animals, blood urea nitrogen was 65 +/- 21, 166 +/- 28, and 50 +/- 14 mg/dl (P = 0.002); creatinine was 0.14 +/- 0.05, 0.74 +/- 0.26, and 0.23 +/- 0.27 mg/dl (P = 0.014); necrotic tubules were 2.5 +/- 0.25%, 12.0 +/- 1.9%, and 5.0 +/- 1.6% (P = 0.004), respectively. In ER-[alpha] and ER-[beta] gene-deleted mice and controls estradiol-reduced functional injury (blood urea nitrogen: estradiol 117 +/- 71, vehicle 167 +/- 56, P = 0.007; creatinine: estradiol 0.5 +/- 0.5, vehicle 1.0 +/- 0.4, P = 0.013), but the effect of estradiol was not different between ER-[alpha] or ER-[beta] gene-deleted mice. Adding ICI 182,780 to estradiol did not alter injury. CONCLUSIONS In women, kidneys were protected from cardiac arrest through estrogen. Estradiol-mediated renoprotection was not affected by ER deletion or blockade. Estradiol is renoprotective after cardiac arrest. The results indicate that estradiol renoprotection is ER-[alpha] and ER-[beta] independent.
Collapse
|
134
|
Rybalchenko V, Grillo MA, Gastinger MJ, Rybalchenko N, Payne AJ, Koulen P. The unliganded long isoform of estrogen receptor beta stimulates brain ryanodine receptor single channel activity alongside with cytosolic Ca2+. J Recept Signal Transduct Res 2010; 29:326-41. [PMID: 19899956 DOI: 10.3109/10799890903295168] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ca(2+) release from intracellular stores mediated by endoplasmic reticulum membrane ryanodine receptors (RyR) plays a key role in activating and synchronizing downstream Ca(2+)-dependent mechanisms, in different cells varying from apoptosis to nuclear transcription and development of defensive responses. Recently discovered, atypical "nongenomic" effects mediated by estrogen receptors (ER) include rapid Ca(2+) release upon estrogen exposure in conditions implicitly suggesting involvement of RyRs. In the present study, we report various levels of colocalization between RyR type 2 (RyR2) and ER type beta (ER beta) in the neuronal cell line HT-22, indicating a possible functional interaction. Electrophysiological analyses revealed a significant increase in single-channel ionic currents generated by mouse brain RyRs after application of the soluble monomer of the long form ER beta (ER beta 1). The effect was due to a strong increase in open probability of RyR higher open channel sublevels at cytosolic [Ca(2+)] concentrations of 100 nM, suggesting a synergistic action of ER beta 1 and Ca(2+) in RyR activation, and a potential contribution to Ca(2+)-induced Ca(2+) release rather than to basal intracellular Ca(2+) concentration level at rest. This RyR/ER beta interaction has potential effects on cellular physiology, including roles of shorter ER beta isoforms and modulation of the RyR/ER beta complexes by exogenous estrogens.
Collapse
|
135
|
Henderson VW, Brinton RD. Menopause and mitochondria: windows into estrogen effects on Alzheimer's disease risk and therapy. PROGRESS IN BRAIN RESEARCH 2010; 182:77-96. [PMID: 20541661 PMCID: PMC5776041 DOI: 10.1016/s0079-6123(10)82003-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Metabolic derangements and oxidative stress are early events in Alzheimer's disease pathogenesis. Multi-faceted effects of estrogens include improved cerebral metabolic profile and reduced oxidative stress through actions on mitochondria, suggesting that a woman's endogenous and exogenous estrogen exposures during midlife and in the late post-menopause might favourably influence Alzheimer risk and symptoms. This prediction finds partial support in the clinical literature. As expected, early menopause induced by oophorectomy may increase cognitive vulnerability; however, there is no clear link between age at menopause and Alzheimer risk in other settings, or between natural menopause and memory loss. Further, among older post-menopausal women, initiating estrogen-containing hormone therapy increases dementia risk and probably does not improve Alzheimer's disease symptoms. As suggested by the 'critical window' or 'healthy cell' hypothesis, better outcomes might be expected from earlier estrogen exposures. Some observational results imply that effects of hormone therapy on Alzheimer risk are indeed modified by age at initiation, temporal proximity to menopause, or a woman's health. However, potential methodological biases warrant caution in interpreting observational findings. Anticipated results from large, ongoing clinical trials [Early Versus Late Intervention Trial with Estradiol (ELITE), Kronos Early Estrogen Prevention Study (KEEPS)] will help settle whether midlife estrogen therapy improves midlife cognitive skills but not whether midlife estrogen exposures modify late-life Alzheimer risk. Estrogen effects on mitochondria adumbrate the potential relevance of estrogens to Alzheimer's disease. However, laboratory models are inexact embodiments of Alzheimer pathogenesis and progression, making it difficult to surmise net effects of estrogen exposures. Research needs include better predictors of adverse cognitive outcomes, biomarkers for risks associated with hormone therapy, and tools for monitoring brain function and disease progression.
Collapse
Affiliation(s)
- Victor W Henderson
- Department of Health Research & Policy (Epidemiology), Stanford University, Stanford, CA, USA.
| | | |
Collapse
|
136
|
Developmental Profile and Subcellular Localization of The Non-genomic Membranous Estrogen Receptor GPR30 in The Hippocampus of Postnatal Female Rats*. PROG BIOCHEM BIOPHYS 2009. [DOI: 10.3724/sp.j.1206.2008.00207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
137
|
Grassi S, Frondaroli A, Scarduzio M, Dutia MB, Dieni C, Pettorossi VE. Effects of 17beta-estradiol on glutamate synaptic transmission and neuronal excitability in the rat medial vestibular nuclei. Neuroscience 2009; 165:1100-14. [PMID: 19944747 DOI: 10.1016/j.neuroscience.2009.11.039] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 11/17/2009] [Indexed: 10/20/2022]
Abstract
We investigated the effects of the neurosteroid 17beta-estradiol (E(2)) on the evoked and spontaneous activity of rat medial vestibular nucleus (MVN) neurons in brainstem slices. E(2) enhances the synaptic response to vestibular nerve stimulation in type B neurons and depresses the spontaneous discharge in both type A and B neurons. The amplitude of the field potential, as well as the excitatory post-synaptic potential (EPSP) and current (EPSC), in type B neurons, are enhanced by E(2). Both effects are long-term phenomena since they outlast the drug washout. The enhancement of synaptic response is mainly due to facilitation of glutamate release mediated by pre-synaptic N-methyl-D-aspartate receptors (NMDARs), since the reduction of paired pulse ratio (PPR) and the increase of miniature EPSC frequency after E(2) are abolished under D-(-)-2-amino-5-phosphonopentanoic acid (AP-5). E(2) also facilitates post-synaptic NMDARs, but it does not affect directly alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) and group I-metabotropic glutamate receptors (mGluRs-I). In contrast, the depression of the spontaneous discharge of type A and type B neurons appears to depend on E(2) modulation of intrinsic ion conductances, as the effect remains after blockade of glutamate, GABA and glycine receptors (GlyRs). The net effect of E(2) is to enhance the signal-to-noise ratio of the synaptic response in type B neurons, relative to resting activity of all MVN neurons. These findings provide evidence for a novel potential mechanism to modulate the responsiveness of vestibular neurons to afferent inputs, and so regulate vestibular function in vivo.
Collapse
Affiliation(s)
- S Grassi
- Department of Internal Medicine, Section of Human Physiology, University of Perugia, Perugia, Italy.
| | | | | | | | | | | |
Collapse
|
138
|
Ramírez CM, González M, Díaz M, Alonso R, Ferrer I, Santpere G, Puig B, Meyer G, Marin R. VDAC and ERα interaction in caveolae from human cortex is altered in Alzheimer's disease. Mol Cell Neurosci 2009; 42:172-83. [DOI: 10.1016/j.mcn.2009.07.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 06/23/2009] [Accepted: 07/01/2009] [Indexed: 10/20/2022] Open
|
139
|
Ghosh S, Thakur MK. Interaction of estrogen receptor-alpha ligand binding domain with nuclear proteins of aging mouse brain. J Neurosci Res 2009; 87:2591-600. [PMID: 19326447 DOI: 10.1002/jnr.22068] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
After the interaction of estrogen with the ligand binding domain (LBD) of mouse estrogen receptor-alpha (mERalpha) and hormone-responsive elements of target genes, many nuclear proteins are recruited to regulate the expression of specific genes. Because it is not known which brain proteins interact with LBD or whether these proteins vary with age and sex, we used pull-down assay and far Western blotting to detect five nuclear proteins of 160, 140, 87, 60, and 46 kD in the mouse brain. These interacting proteins were identified as PELP1, RIP140, PGC1alpha, BAF60, and ADA3, respectively. The level of PELP1, RIP140, PGC1alpha, and BAF60 decreased drastically in old compared with adult male mice, whereas the ADA3 level showed no significant change. PELP1, PGC1alpha, and BAF60 levels were lower in old male compared with female mice. Thus we report the identification and interaction of five nuclear proteins with mERalpha-LBD, indicating their role in estrogen signaling and brain functions during aging.
Collapse
Affiliation(s)
- Swati Ghosh
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
140
|
Long-term potentiation in the rat medial vestibular nuclei depends on locally synthesized 17beta-estradiol. J Neurosci 2009; 29:10779-83. [PMID: 19710328 DOI: 10.1523/jneurosci.1697-09.2009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In male rat brainstem slices, we investigated the involvement of locally synthesized 17beta-estradiol (E(2)) in the induction in the medial vestibular nucleus (MVN) of long-term potentiation (LTP) by high-frequency stimulation (HFS) of the primary vestibular afferents. We demonstrated that the blockade of aromatase by letrozole or of E(2) receptors (ERalpha and ERbeta) by ICI 182,780 prevented the HFS-induced LTP of the N1 wave of the evoked field potential (FP) without affecting baseline responses. Only prolonged afferent activation could induce low LTP. In contrast, HFS applied under a combined blockade of GABA(A) receptors and aromatase or ERs was still able to induce LTP, but it was significantly lower and slower. These findings demonstrate that E(2) does not have a tonic influence on the activity of the MVN neurons and provide the first evidence of the crucial role played by local synthesis of E(2) in inducing LTP. We suggest that the synthesis of E(2) occurs after aromatase activation during HFS and facilitates the development of vestibular synaptic plasticity by influencing glutamate and GABA transmission.
Collapse
|
141
|
Lebesgue D, Reyna-Neyra A, Huang X, Etgen AM. GPR30 differentially regulates short latency responses of luteinising hormone and prolactin secretion to oestradiol. J Neuroendocrinol 2009; 21:743-52. [PMID: 19523168 PMCID: PMC2756443 DOI: 10.1111/j.1365-2826.2009.01893.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Rapid, nongenomic actions of 17beta-oestradiol (E(2)) on hypothalamic neurones that may be relevant to reproductive function were described decades ago. The orphan G protein-coupled receptor, GPR30, was recently shown to bind oestrogens and to trigger rapid signalling in vitro, and is expressed in several rat and human brain regions, including the hypothalamus. We used two complementary approaches to investigate the role of GPR30 in hypothalamic responses to E(2) that are relevant to reproductive physiology. Serial blood sampling after the acute administration of the selective GPR30 agonist G1 was used to assess the role of GPR30 in short latency negative-feedback inhibition of luteinising hormone (LH) secretion and facilitation of prolactin secretion in ovariohysterectomised female rats. In vivo RNA interference (RNAi), mediated by adeno-associated virus-expressing small hairpin RNA (shRNA) infused into the mediobasal hypothalamus, was used to study the effects of GPR30 knockdown on these rapid responses to E(2). Longer-term actions of E(2) on female sexual behaviour (lordosis) were also examined in female rats subjected to in vivo RNAi. Administration of E(2) or G1 triggered a short latency surge of prolactin secretion, and animals subjected to GPR30 RNAi showed significantly less E(2)-dependent prolactin release than animals receiving control virus. G1 did not mimic E(2) negative-feedback inhibition of LH secretion, and GPR30 RNAi did not interfere with E(2) suppression of LH or facilitation of lordosis behaviour. These findings suggest that activation of GPR30 promotes short latency prolactin secretion but does not mediate E(2) negative-feedback inhibition of LH secretion or E(2) facilitation of female reproductive behaviour.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cells, Cultured
- Estradiol/administration & dosage
- Estradiol/pharmacology
- Female
- Humans
- Injections, Intraventricular
- Luteinizing Hormone/blood
- Luteinizing Hormone/metabolism
- Posture/physiology
- Prolactin/blood
- Prolactin/metabolism
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- RNA, Small Interfering/pharmacology
- Rats
- Rats, Sprague-Dawley
- Rats, Transgenic
- Reaction Time/drug effects
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/physiology
- Sexual Behavior, Animal/drug effects
- Time Factors
- Transduction, Genetic
Collapse
Affiliation(s)
- D Lebesgue
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | |
Collapse
|
142
|
Micevych P, Dominguez R. Membrane estradiol signaling in the brain. Front Neuroendocrinol 2009; 30:315-27. [PMID: 19416735 PMCID: PMC2720427 DOI: 10.1016/j.yfrne.2009.04.011] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 04/22/2009] [Accepted: 04/23/2009] [Indexed: 12/16/2022]
Abstract
While the physiology of membrane-initiated estradiol signaling in the nervous system has remained elusive, a great deal of progress has been made toward understanding the activation of cell signaling. Membrane-initiated estradiol signaling activates G proteins and their downstream cascades, but the identity of membrane receptors and the proximal signaling mechanism(s) have been more difficult to elucidate. Mounting evidence suggests that classical intracellular estrogen receptor-alpha (ERalpha) and ERbeta are trafficked to the membrane to mediate estradiol cell signaling. Moreover, an interaction of membrane ERalpha and ERbeta with metabotropic glutamate receptors has been identified that explains the pleomorphic actions of membrane-initiated estradiol signaling. This review focuses on the mechanism of actions initiated by membrane estradiol receptors and discusses the role of scaffold proteins and signaling cascades involved in the regulation of nociception, sexual receptivity and the synthesis of neuroprogesterone, an important component in the central nervous system signaling.
Collapse
Affiliation(s)
- Paul Micevych
- Department of Neurobiology and the Laboratory of Neuroendocrinology David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1763, USA.
| | | |
Collapse
|
143
|
|
144
|
Lebesgue D, Chevaleyre V, Zukin RS, Etgen AM. Estradiol rescues neurons from global ischemia-induced cell death: multiple cellular pathways of neuroprotection. Steroids 2009; 74:555-61. [PMID: 19428444 PMCID: PMC3029071 DOI: 10.1016/j.steroids.2009.01.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 01/08/2009] [Accepted: 01/08/2009] [Indexed: 02/03/2023]
Abstract
The potential neuroprotective role of sex hormones in chronic neurodegenerative disorders and acute brain ischemia following cardiac arrest and stroke is of a great therapeutic interest. Long-term pretreatment with estradiol and other estrogens affords robust neuroprotection in male and female rodents subjected to focal and global ischemia. However, the receptors (e.g., cell surface or nuclear), intracellular signaling pathways and networks of estrogen-regulated genes that intervene in neuronal apoptosis are as yet unclear. We have shown that estradiol administered at physiological levels for two weeks before ischemia rescues neurons destined to die in the hippocampal CA1 and ameliorates ischemia-induced cognitive deficits in ovariectomized female rats. This regimen of estradiol treatment involves classical intracellular estrogen receptors, transactivation of IGF-1 receptors and stimulation of the ERK/MAPK signaling pathway, which in turn maintains CREB activity in the ischemic CA1. We also find that a single, acute injection of estradiol administrated into the brain ventricle immediately after an ischemic event reduces both neuronal death and cognitive deficits. Because these findings suggest that hormones could be used to treat patients when given after brain ischemia, it is critical to determine whether the same or different pathways mediate this form of neuroprotection. We find that an agonist of the membrane estrogen receptor GPR30 mimics short latency estradiol facilitation of synaptic transmission in the hippocampus. Therefore, we are testing the hypothesis that GPR30 may act together with intracellular estrogen receptors to activate cell signaling pathways to promote neuron survival after global ischemia.
Collapse
Affiliation(s)
- Diane Lebesgue
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | | | |
Collapse
|
145
|
Bourque M, Dluzen DE, Di Paolo T. Neuroprotective actions of sex steroids in Parkinson's disease. Front Neuroendocrinol 2009; 30:142-57. [PMID: 19410597 DOI: 10.1016/j.yfrne.2009.04.014] [Citation(s) in RCA: 181] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 04/22/2009] [Accepted: 04/23/2009] [Indexed: 12/16/2022]
Abstract
The sex difference in Parkinson's disease, with a higher susceptibility in men, suggests a modulatory effect of sex steroids in the brain. Numerous studies highlight that sex steroids have neuroprotective properties against various brain injuries. This paper reviews the protective effects of sex hormones, particularly estradiol, progesterone and androgens, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) animal model of Parkinson's disease as compared to methamphetamine toxicity. The molecular mechanisms underlying beneficial actions of sex steroids on the brain have been investigated showing steroid, dose, timing and duration specificities and presently focus is on the dopamine signaling pathways, the next frontier. Both genomic and non-genomic actions of estrogen converge to promote survival factors and show sex differences. Neuroprotection by estrogen involves activation of signaling molecules such as the phosphatidylinositol-3 kinase/Akt and the mitogen-activated protein kinase pathways. Interaction with growth factors, such as insulin-like growth factor 1, also contributes to protective actions of estrogen.
Collapse
Affiliation(s)
- Mélanie Bourque
- Molecular Endocrinology and Genomic Research Center, Laval University Medical Center, CHUL, Quebec City, Quebec, Canada
| | | | | |
Collapse
|
146
|
Suzuki S, Brown CM, Wise PM. Neuroprotective effects of estrogens following ischemic stroke. Front Neuroendocrinol 2009; 30:201-11. [PMID: 19401209 PMCID: PMC3672220 DOI: 10.1016/j.yfrne.2009.04.007] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2009] [Revised: 04/15/2009] [Accepted: 04/17/2009] [Indexed: 02/07/2023]
Abstract
Our laboratory has investigated whether and how 17beta-estradiol (E(2)) protects the brain against neurodegeneration associated with cerebrovascular stroke. We have discovered that low, physiological concentrations of E(2), which are strikingly similar to low-basal circulating levels found in cycling mice, dramatically protect the brain against stroke injury, and consequently revealed multiple signaling pathways and key genes that mediate protective action of E(2). Here we will review the discoveries comprising our current understanding of neuroprotective actions of estrogens against ischemic stroke. These findings may carry far reaching implications for improving the quality of life in aging populations.
Collapse
Affiliation(s)
- Shotaro Suzuki
- Departments of Physiology and Biophysics, and Biology, University of Washington, Seattle, WA 98195-1237, USA
| | | | | |
Collapse
|
147
|
Arnold S, Beyer C. Neuroprotection by estrogen in the brain: the mitochondrial compartment as presumed therapeutic target. J Neurochem 2009; 110:1-11. [DOI: 10.1111/j.1471-4159.2009.06133.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
148
|
Kipp M, Beyer C. Impact of sex steroids on neuroinflammatory processes and experimental multiple sclerosis. Front Neuroendocrinol 2009; 30:188-200. [PMID: 19393685 DOI: 10.1016/j.yfrne.2009.04.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 04/01/2009] [Accepted: 04/14/2009] [Indexed: 12/18/2022]
Abstract
Synthetic and natural estrogens as well as progestins modulate neuronal development and activity. Neurons and glia are endowed with high-affinity steroid receptors. Besides regulating brain physiology, both steroids conciliate neuroprotection against toxicity and neurodegeneration. The majority of data derive from in vitro studies, although more recently, animal models have proven the efficaciousness of steroids as neuroprotective factors. Indications for a safeguarding role also emerge from first clinical trials. Gender-specific prevalence of degenerative disorders might be associated with the loss of hormonal activity or steroid malfunctions. Our studies and evidence from the literature support the view that steroids attenuate neuroinflammation by reducing the pro-inflammatory property of astrocytes. This effect appears variable depending on the brain region and toxic condition. Both hormones can individually mediate protection, but they are more effective in cooperation. A second research line, using an animal model for multiple sclerosis, provides evidence that steroids achieve remyelination after demyelination. The underlying cellular mechanisms involve interactions with astroglia, insulin-like growth factor-1 responses, and the recruitment of oligodendrocytes.
Collapse
Affiliation(s)
- Markus Kipp
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
149
|
Miranda JA, Liu RC. Dissecting natural sensory plasticity: hormones and experience in a maternal context. Hear Res 2009; 252:21-8. [PMID: 19401225 PMCID: PMC2698041 DOI: 10.1016/j.heares.2009.04.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2008] [Revised: 04/04/2009] [Accepted: 04/12/2009] [Indexed: 10/20/2022]
Abstract
There is a growing consensus that the auditory system is dynamic in its representation of behaviorally relevant sounds. The auditory cortex in particular seems to be an important locus for plasticity that may reflect the memory of such sounds, or functionally improve their processing. The mechanisms that underlie these changes may be either intrinsic because they depend on the receiver's physiological state, or extrinsic because they arise from the context in which behavioral relevance is gained. Research in a mouse model of acoustic communication between offspring and adult females offers the opportunity to explore both of these contributions to auditory cortical plasticity in a natural context. Recent works have found that after the vocalizations of infant mice become behaviorally relevant to mothers, auditory cortical activity is significantly changed in a way that may improve their processing. Here we consider the hypothesis that maternal hormones (intrinsic factor) and sensory experience (extrinsic factor) contribute together to drive these changes, focusing specifically on the evidence that well-known experience-dependent mechanisms of cortical plasticity can be modulated by hormones.
Collapse
Affiliation(s)
- Jason A. Miranda
- Department of Biology, Emory University, 1510 Clifton Road, Room 2006, Atlanta, GA 30322, USA,
- Center for Behavioral Neuroscience, PO Box 3966, Atlanta, GA 30302, USA,
| | - Robert C. Liu
- Department of Biology, Emory University, 1510 Clifton Road, Room 2006, Atlanta, GA 30322, USA,
- Center for Behavioral Neuroscience, PO Box 3966, Atlanta, GA 30302, USA,
| |
Collapse
|
150
|
Graziottin A, Serafini A. Depression and the menopause: why antidepressants are not enough? ACTA ACUST UNITED AC 2009; 15:76-81. [DOI: 10.1258/mi.2009.009021] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Gender differences, related to varying sexual hormone levels and hormone secretion patterns across the lifespan, contribute to women's vulnerability to mood disorders and major depression. Women are more prone than men to depression, from puberty onwards, with a specific exposure across the menopausal transition. However, controversy still exists in considering fluctuation/loss of estrogen as a specific aetiologic factor contributing to depression in perimenopause and beyond. Aims To briefly review the interaction between changes in menopausal hormone levels, mood disorders, associated neuropsychological co-morbidities and ageing, and to evaluate the currently available therapeutic options for perimenopausal mood disorders: (a) treatment of light to moderate mood disorders with hormonal therapy (HT); (b) treatment of major depression with antidepressants; (c) the synergistic effect between HT and antidepressants in treating menopausal depression. Results Depression across the menopause has a multifactorial aetiology. Predictive factors include: previous depressive episodes such as premenstrual syndrome and/or postpartum depression; co-morbidity with major menopausal symptoms, especially hot flashes, nocturnal sweating, insomnia; menopause not treated with HT; major existential stress; elevated body mass index; low socioeconomic level and ethnicity. Postmenopausal depression is more severe, has a more insidious course, is more resistant to conventional antidepressants in comparison with premenopausal women and has better outcomes when antidepressants are combined with HT. Conclusion The current evidence contributes to a re-reading of the relationship between menopause and depression. The combination of the antidepressant with HT seems to offer the best therapeutic potential in terms of efficacy, rapidity of improvement and consistency of remission in the follow-up.
Collapse
Affiliation(s)
| | - Audrey Serafini
- IRCCS San Raffaele Department of Obstetrics and Gynecology, Milan, Italy
| |
Collapse
|