1
|
Puga-Olguín A, Rodríguez-Landa JF, Rovirosa-Hernández MDJ, Germán-Ponciano LJ, Caba M, Meza E, Guillén-Ruiz G, Olmos-Vázquez OJ. Long-term ovariectomy increases anxiety- and despair-like behaviors associated with lower Fos immunoreactivity in the lateral septal nucleus in rats. Behav Brain Res 2019; 360:185-195. [DOI: 10.1016/j.bbr.2018.12.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/24/2018] [Accepted: 12/06/2018] [Indexed: 01/01/2023]
|
2
|
Affiliation(s)
- Bruce Becker
- Department of Emergency Medicine, Division of Sex and Gender in Emergency Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Alyson J. McGregor
- Department of Emergency Medicine, Division of Sex and Gender in Emergency Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
3
|
|
4
|
Pedrón VT, Taravini IR, Induni AS, Balerio GN. Baclofen did not modify sexually dimorphic c-Fos expression during morphine withdrawal syndrome. Synapse 2012; 67:118-26. [DOI: 10.1002/syn.21620] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 10/26/2012] [Indexed: 12/18/2022]
|
5
|
Palmeira CCDA, Ashmawi HA, Posso IDP. Sex and pain perception and analgesia. Rev Bras Anestesiol 2012; 61:814-28. [PMID: 22063383 DOI: 10.1016/s0034-7094(11)70091-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 02/21/2011] [Indexed: 10/26/2022] Open
Abstract
Sex is an important factor in painful experience modulation. Large volume of evidence shows that experience is different for males and females, as well as the answer to some classes of analgesics. Laboratory experiments suggest that women have a lower pain threshold than men related to pain from noxious stimuli such as heat, cold, pressure and electrical stimulation. Pain is a dynamic phenomenon under the influence of various mechanisms of excitatory and inhibitory control. The differences in pain perception related to sex may be associated with hyperalgesia in women, but also to the hypoactivity of the inhibitory system of pain in females. The purpose of this review besides showing some relationship for gonadal hormones, central nervous system and pain is to provide reference points for the discussion of one of the most intriguing aspects of the pathophysiology of pain: the differences in the presence of painful stimuli related to gender.
Collapse
|
6
|
Nisenblat V, Engel-Yeger B, Ohel G, Aronson D, Granot M. The association between supra-physiological levels of estradiol and response patterns to experimental pain. Eur J Pain 2012; 14:840-6. [DOI: 10.1016/j.ejpain.2010.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 12/31/2009] [Accepted: 01/16/2010] [Indexed: 10/19/2022]
|
7
|
Buchanan F, Myles P, Cicuttini F. Effect of patient sex on general anaesthesia and recovery. Br J Anaesth 2011; 106:832-9. [DOI: 10.1093/bja/aer094] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
8
|
|
9
|
Pleil KE, Glenn MJ, Williams CL. Estradiol alters Fos-immunoreactivity in the hippocampus and dorsal striatum during place and response learning in middle-aged but not young adult female rats. Endocrinology 2011; 152:946-56. [PMID: 21285311 PMCID: PMC3040062 DOI: 10.1210/en.2010-0715] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 12/08/2010] [Indexed: 11/19/2022]
Abstract
Evidence from lesion and inactivation studies suggests that the hippocampus (HPC) and dorsal striatum compete for control over navigation behavior, and there is some evidence in males that the structure with greater relative activation controls behavior. Estradiol has been shown to enhance HPC-dependent place learning and impair dorsal striatum-dependent response learning in female rats, possibly by increasing hippocampal activation and/or decreasing striatal activation. We used Fos-immunoreactivity (Fos-IR) to examine the activation of several subregions of the HPC and striatum in ovariectomized female rats with or without estradiol replacement 30 min after place or response learning. In 4-month-old rats, neither task nor estradiol increased Fos-IR above explore control levels in any subregion analyzed, even though estradiol impaired response learning. In 12-month-old rats, estradiol increased Fos-IR in the dentate gyrus, dorsal medial striatum, and dorsal lateral striatum in place task learners, while the absence of estradiol increased Fos-IR in these regions in response task learners. However, learning rate was not affected by estradiol in either task. We also included a group of long-term ovariectomized 12-month-old rats that displayed impaired place learning and altered Fos-IR in CA1 of the HPC. These results suggest that task-specific effects of estradiol on hippocampal and striatal activation emerge across age but that relative hippocampal and striatal activation are not related to learning rate during spatial navigation learning.
Collapse
Affiliation(s)
- Kristen E Pleil
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina 27708, USA
| | | | | |
Collapse
|
10
|
Wu TW, Chen S, Brinton RD. Membrane estrogen receptors mediate calcium signaling and MAP kinase activation in individual hippocampal neurons. Brain Res 2011; 1379:34-43. [PMID: 21241678 DOI: 10.1016/j.brainres.2011.01.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2011] [Indexed: 11/19/2022]
Abstract
Previously we demonstrated that 17β-Estradiol (E2) induced rapid Ca(2+) influx via L-type calcium channel activation, which was required for activation of Src/ERK/CREB/Bcl2 signaling cascade and subsequent induction of neuroprotective and neurotrophic responses in rat hippocampal and cortical neurons (Wu et al., 2005; Zhao et al., 2005). The current study determined the presence and specificity of membrane E2 binding sites and the functional consequence of E2 binding to membrane receptors in individual neurons. Using E2-BSA-FITC (fluorescein isothiocyanate) macromolecular complex, membrane E2 binding sites were observed in hippocampal neurons. Punctate FITC signal was observed on plasma membrane of soma and neuronal processes in E2-BSA-FITC binding neurons. No membrane binding was observed with BSA-FITC. Specificity of binding was demonstrated by competition with excess un-conjugated E2. An ERa specific agonist, PPT, and an ERb agonist, DPN, partially competed for E2-BSA-FITC binding. Imaging of intracellular Ca(2+) ([Ca(2+)]i) in live neurons, revealed rapid Ca(2+) responses in E2-BSA-FITC binding neurons within minutes that culminated in a greater [Ca(2+)]i rise and [Ca(2+)]i spikes at >20 min. The same neurons in which E2-BSA-FITC induced a [Ca(2+)]i rise also exhibited activated pERK (extracellular signal-regulated kinase) that was translocated to the nucleus. Immunofluorescent analyses demonstrated that both excitatory and inhibitory neuronal markers labeled subpopulations of E2-BSA-FITC binding neurons. All E2-BSA-FITC binding neurons expressed L-type calcium channels. These results demonstrate, at a single cell level, that E2 membrane receptors mediate the rapid signaling cascades required for E2 neuroprotective and neurotrophic effects in hippocampal neurons. These results are discussed with respect to therapeutic targets of estrogen therapy in brain.
Collapse
Affiliation(s)
- Tzu-Wei Wu
- University of Southern California, Pharmaceutical Sciences Center, Los Angeles, CA 90089-9121, USA.
| | | | | |
Collapse
|
11
|
Walf AA, Paris JJ, Rhodes ME, Simpkins JW, Frye CA. Divergent mechanisms for trophic actions of estrogens in the brain and peripheral tissues. Brain Res 2010; 1379:119-36. [PMID: 21130078 DOI: 10.1016/j.brainres.2010.11.081] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 11/23/2010] [Accepted: 11/23/2010] [Indexed: 01/08/2023]
Abstract
17β-estradiol (E(2)) can enhance reproductive, cognitive, and affective functions; however, the mechanisms by which E(2) has these effects need to be better understood. Pleiotrophic effects of E(2) can occur via traditional and novel actions at various forms of estrogen receptors (ERs). In the central nervous system, trophic effects of E(2) may be related to beneficial effects of hormone replacement therapy (HRT). However, in peripheral reproductive tissues, E(2)'s capacity to evoke growth can increase risk of cancers. This review focuses on investigations aimed at elucidating divergent mechanisms of steroids to promote trophic effects in the brain, independent of effects on peripheral reproductive tissues. First, actions of estrogens via ERα or ERβ for peripheral growth (carcinogen-induced tumors, uterine growth) and hippocampus-dependent behaviors (affect, cognition) are described. Second, factors that influence these effects of estrogens are described (e.g. experience, timing/critical windows, non-ER mechanisms). Third, effects of estrogens at ERβ related to actions of progestogens, such as 5α-pregnan-3α-ol-20-one (3α,5α-THP) are described. In summary, effects of E(2) may occur via multiple mechanisms, which may underlie favorable effects in the brain with minimal peripheral trophic effects.
Collapse
Affiliation(s)
- Alicia A Walf
- Life Sciences Research, University at Albany, Albany, NY 12222, USA
| | | | | | | | | |
Collapse
|
12
|
Aloisi AM, Ceccarelli I, Fiorenzani P, Maddalena M, Rossi A, Tomei V, Sorda G, Danielli B, Rovini M, Cappelli A, Anzini M, Giordano A. Aromatase and 5-alpha reductase gene expression: modulation by pain and morphine treatment in male rats. Mol Pain 2010; 6:69. [PMID: 20977699 PMCID: PMC2978140 DOI: 10.1186/1744-8069-6-69] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 10/26/2010] [Indexed: 01/18/2023] Open
Abstract
Background The steroid hormone testosterone has been found to be greatly reduced by opioids in different experimental and clinical conditions. The purpose of this study on male rats was to determine the effects of a single injection of morphine (5 mg/Kg) on persistent pain (formalin test) and the single or combined effects on p450-aromatase and 5-alpha reductase type 1 mRNA expression in the brain, liver and testis. Testosterone was determined in the plasma and in the brain, morphine was assayed in the plasma. Results In the morphine-treated rats, there were increases of 5-alpha reductase mRNA expression in the liver and aromatase mRNA expression in the brain and gonads. Morphine was detected in the blood of all morphine-treated rats even though there were no clear analgesic affects in the formalin-treated animals three hours after treatment. Testosterone was greatly reduced in the plasma and brain in morphine-treated subjects. Conclusions It appears that morphine administration can induce long-lasting genomic effects in different body areas which contribute to the strong central and peripheral testosterone levels. These changes were not always accompanied by behavioral modifications.
Collapse
Affiliation(s)
- Anna Maria Aloisi
- Department of Physiology, Neuroscience and Applied Physiology Unit, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
McEwen BS. Stress, sex, and neural adaptation to a changing environment: mechanisms of neuronal remodeling. Ann N Y Acad Sci 2010; 1204 Suppl:E38-59. [PMID: 20840167 PMCID: PMC2946089 DOI: 10.1111/j.1749-6632.2010.05568.x] [Citation(s) in RCA: 241] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The adult brain is much more resilient and adaptable than previously believed, and adaptive structural plasticity involves growth and shrinkage of dendritic trees, turnover of synapses, and limited amounts of neurogenesis in the forebrain, especially the dentate gyrus of the hippocampal formation. Stress and sex hormones help to mediate adaptive structural plasticity, which has been extensively investigated in the hippocampus and to a lesser extent in the prefrontal cortex and amygdala, all brain regions that are involved in cognitive and emotional functions. Stress and sex hormones exert their effects on brain structural remodeling through both classical genomic as well as non-genomic mechanisms, and they do so in collaboration with neurotransmitters and other intra- and extracellular mediators. This review will illustrate the actions of estrogen on synapse formation in the hippocampus and the process of stress-induced remodeling of dendrites and synapses in the hippocampus, amygdala, and prefrontal cortex. The influence of early developmental epigenetic events, such as early life stress and brain sexual differentiation, is noted along with the interactions between sex hormones and the effects of stress on the brain. Because hormones influence brain structure and function and because hormone secretion is governed by the brain, applied molecular neuroscience techniques can begin to reveal the role of hormones in brain-related disorders and the treatment of these diseases. A better understanding of hormone-brain interactions should promote more flexible approaches to the treatment of psychiatric disorders, as well as their prevention through both behavioral and pharmaceutical interventions.
Collapse
Affiliation(s)
- Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
14
|
Ceccarelli I, Fiorenzani P, Della Seta D, Massafra C, Cinci G, Bocci A, Aloisi AM. Perinatal exposure to xenoestrogens affects pain in adult female rats. Neurotoxicol Teratol 2009; 31:203-9. [DOI: 10.1016/j.ntt.2009.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Revised: 01/27/2009] [Accepted: 02/16/2009] [Indexed: 11/25/2022]
|
15
|
Buchanan FF, Myles PS, Cicuttini F. Patient Sex and its Influence on General Anaesthesia. Anaesth Intensive Care 2009; 37:207-18. [DOI: 10.1177/0310057x0903700201] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Physiological and pharmacological differences exist between men and women. Women wake faster than men following general anaesthesia. Women also differ from men in their postoperative recovery as reflected by differences in postoperative pain, nausea and vomiting and overall quality of recovery. These gender differences seem to be more pronounced in premenopausal women, suggesting hormonal mechanisms are a major contributing factor.
Collapse
Affiliation(s)
- F. F. Buchanan
- Department of Anaesthesia and Perioperative Medicine, Alfred Hospital and Academic Board of Anaesthesia and Perioperative Medicine, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Anaesthesia and Perioperative Medicine, Alfred Hospital
| | - P. S. Myles
- Department of Anaesthesia and Perioperative Medicine, Alfred Hospital and Academic Board of Anaesthesia and Perioperative Medicine, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Anaesthesia and Perioperative Medicine, Alfred Hospital, Academic Board of Anaesthesia and Perioperative Medicine, Monash University and NHMRC Practitioner Fellow
| | - F. Cicuttini
- Department of Anaesthesia and Perioperative Medicine, Alfred Hospital and Academic Board of Anaesthesia and Perioperative Medicine, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Rheumatology Unit, Alfred Hospital and Department of Epidemiology and Monash University
| |
Collapse
|
16
|
Scharfman HE, Hintz TM, Gomez J, Stormes KA, Barouk S, Malthankar-Phatak GH, McCloskey DP, Luine VN, Maclusky NJ. Changes in hippocampal function of ovariectomized rats after sequential low doses of estradiol to simulate the preovulatory estrogen surge. Eur J Neurosci 2007; 26:2595-612. [PMID: 17970745 DOI: 10.1111/j.1460-9568.2007.05848.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In adult female rats, robust hippocampal changes occur when estradiol rises on the morning of proestrus. Whether estradiol mediates these changes, however, remains unknown. To address this issue, we used sequential injections of estradiol to simulate two key components of the preovulatory surge: the rapid rise in estradiol on proestrous morning, and the slower rise during the preceding day, diestrus 2. Animals were examined mid-morning of simulated proestrus, and compared to vehicle-treated or intact rats. In both simulated and intact rats, CA1-evoked responses were potentiated in hippocampal slices, and presynaptic mechanisms appeared to contribute. In CA3, multiple population spikes were evoked in response to mossy fiber stimuli, and expression of brain-derived neurotrophic factor was increased. Simulation of proestrous morning also improved performance on object and place recognition tests, in comparison to vehicle treatment. Surprisingly, effects on CA1-evoked responses showed a dependence on estradiol during simulated diestrus 2, as well as a dependence on proestrous morning. Increasing estradiol above the physiological range on proestrous morning paradoxically decreased evoked responses in CA1. However, CA3 pyramidal cell activity increased further, and became synchronized. Together, the results confirm that physiological estradiol levels are sufficient to profoundly affect hippocampal function. In addition: (i) changes on proestrous morning appear to depend on slow increases in estradiol during the preceding day; (ii) effects are extremely sensitive to the peak serum level on proestrous morning; and (iii) there are striking subfield differences within the hippocampus.
Collapse
Affiliation(s)
- Helen E Scharfman
- Center for Neural Recovery and Rehabilitation Research, Helen Hayes Hospital, West Haverstraw, NY 10962, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Mukai H, Tsurugizawa T, Murakami G, Kominami S, Ishii H, Ogiue-Ikeda M, Takata N, Tanabe N, Furukawa A, Hojo Y, Ooishi Y, Morrison JH, Janssen WGM, Rose JA, Chambon P, Kato S, Izumi S, Yamazaki T, Kimoto T, Kawato S. Rapid modulation of long-term depression and spinogenesis via synaptic estrogen receptors in hippocampal principal neurons. J Neurochem 2007; 100:950-67. [PMID: 17266735 DOI: 10.1111/j.1471-4159.2006.04264.x] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Rapid modulation of hippocampal synaptic plasticity by estrogen has long been a hot topic, but analysis of molecular mechanisms via synaptic estrogen receptors has been seriously difficult. Here, two types of independent synaptic plasticity, long-term depression (LTD) and spinogenesis, were investigated, in response to 17beta-estradiol and agonists of estrogen receptors using hippocampal slices from adult male rats. Multi-electrode investigations demonstrated that estradiol rapidly enhanced LTD not only in CA1 but also in CA3 and dentate gyrus. Dendritic spine morphology analysis demonstrated that the density of thin type spines was selectively increased in CA1 pyramidal neurons within 2 h after application of 1 nm estradiol. This enhancement of spinogenesis was completely suppressed by mitogen-activated protein (MAP) kinase inhibitor. Only the estrogen receptor (ER) alpha agonist, (propyl-pyrazole-trinyl)tris-phenol (PPT), induced the same enhancing effect as estradiol on both LTD and spinogenesis in the CA1. The ERbeta agonist, (4-hydroxyphenyl)-propionitrile (DPN), suppressed LTD and did not affect spinogenesis. Because the mode of synaptic modulations by estradiol was mostly the same as that by the ERalpha agonist, a search was made for synaptic ERalpha using purified RC-19 antibody qualified using ERalpha knockout (KO) mice. Localization of ERalpha in spines of principal glutamatergic neurons was demonstrated using immunogold electron microscopy and immunohistochemistry. ERalpha was also located in nuclei, cytoplasm and presynapses.
Collapse
Affiliation(s)
- Hideo Mukai
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba, Meguro, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Murakami G, Tsurugizawa T, Hatanaka Y, Komatsuzaki Y, Tanabe N, Mukai H, Hojo Y, Kominami S, Yamazaki T, Kimoto T, Kawato S. Comparison between basal and apical dendritic spines in estrogen-induced rapid spinogenesis of CA1 principal neurons in the adult hippocampus. Biochem Biophys Res Commun 2006; 351:553-8. [PMID: 17070772 DOI: 10.1016/j.bbrc.2006.10.066] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2006] [Accepted: 10/13/2006] [Indexed: 11/28/2022]
Abstract
Modulation of hippocampal synaptic plasticity by estrogen has been attracting much attention. Here, we demonstrated the rapid effect of 17beta-estradiol on the density and morphology of spines in the stratum oriens (s.o., basal side) and in the stratum lacunosum-moleculare (s.l.m., apical side) by imaging Lucifer Yellow-injected CA1 neurons in adult male rat hippocampal slices, because spines in s.o. and s.l.m. have been poorly understood as compared with spines in the stratum radiatum. The application of 1nM estradiol-induced a rapid increase in the density of spines of pyramidal neurons within 2h. This increase by estradiol was blocked by Erk MAP kinase inhibitor and estrogen receptor inhibitor in both regions. Effect of blockade by agonists of AMPA receptors and NMDA receptors was different between s.o. and s.l.m. In both regions, ERalpha agonist PPT induced the same enhancing effect of spinogenesis as that induced by estradiol.
Collapse
Affiliation(s)
- Gen Murakami
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba 3-8-1, Meguro, Tokyo 153, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mannella P, Brinton RD. Estrogen receptor protein interaction with phosphatidylinositol 3-kinase leads to activation of phosphorylated Akt and extracellular signal-regulated kinase 1/2 in the same population of cortical neurons: a unified mechanism of estrogen action. J Neurosci 2006; 26:9439-47. [PMID: 16971528 PMCID: PMC6674594 DOI: 10.1523/jneurosci.1443-06.2006] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
17Beta-estradiol (E2)-induced neuroprotection is dependent on mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase (PI3K) signaling cascades. We sought to determine whether E2 neuroprotective mechanisms are mediated by a unified signaling cascade activated by estrogen receptor (ER)-PI3K interaction within the same population of neurons or whether E2 activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt are independent signaling events in different neuronal populations. Immunoprecipitation of E2-treated cortical neurons was conducted to determine a protein-protein interaction between ER and the PI3K regulatory subunit p85. Subsequently, cortical neurons were treated with E2 alone or in presence of MAPK inhibitors or PI3K inhibitors. Results of these analyses indicated a protein-protein interaction between ER and p85 that was time-dependent and consistent with the temporal profile for generation of Akt (pAkt) and ERK1/2 phosphorylation (pERK1/2). E2-induced phosphorylation of Akt, was first apparent at 10 min and maximal at 30 min. Simultaneously, E2-induced pERK1/2 was first apparent at 5-10 min and maximal at 30 min. Inhibition of PI3K completely blocked E2 activation of pAkt at 10 and 30 min and blocked E2 activation of ERK1/2 at 10 min, which revealed a PI3K-independent activation of ERK at 30 min. Double immunocytochemical labeling for pERK1/2 and pAkt demonstrated that E2 induced both signaling pathways in the same neurons. These results indicate a unified signaling mechanism for rapid E2 action that leads to the coordinated activation of both pERK1/2 and pAkt in the same population of neurons. Implications of these results for understanding estrogen mechanism of action in neurons and therapeutic development are considered.
Collapse
Affiliation(s)
| | - Roberta Diaz Brinton
- Department of Molecular Pharmacology and Toxicology and
- Program in Neuroscience, University of Southern California, Los Angeles, California 90033
| |
Collapse
|
20
|
Aloisi AM, Bonifazi M. Sex hormones, central nervous system and pain. Horm Behav 2006; 50:1-7. [PMID: 16423353 DOI: 10.1016/j.yhbeh.2005.12.002] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 12/05/2005] [Accepted: 12/06/2005] [Indexed: 01/04/2023]
Abstract
The aim of the present review, which highlights some relationships between sex hormones, the CNS and pain, is to provide reference points for discussion on one of the most intriguing aspects of pain pathophysiology: the presence of sex differences in the response threshold to phasic painful stimuli and in the incidence of chronic pain syndromes. The first part of the review deals with sex steroids and their mechanisms of action. In the second part, the connections between sex steroids, the CNS and pain are illustrated to introduce possible areas of discussion in the study of sex differences in experimental and clinical pain.
Collapse
Affiliation(s)
- Anna Maria Aloisi
- Department of Physiology, Neuroscience and Applied Physiology Section, Polo Scientifico Universitario San Miniato, Via Aldo Moro, 53100 Siena, Italy.
| | | |
Collapse
|
21
|
Walf AA, Frye CA. A review and update of mechanisms of estrogen in the hippocampus and amygdala for anxiety and depression behavior. Neuropsychopharmacology 2006; 31:1097-111. [PMID: 16554740 PMCID: PMC3624621 DOI: 10.1038/sj.npp.1301067] [Citation(s) in RCA: 346] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Estrogen (E2) has many effects in the central nervous system, including effects on anxiety and depression behavior. This review will address effects of E2 on behaviors related to anxiety and depression in women and animal models and include recent findings from our laboratory related to this topic. E2's antianxiety and antidepressant-like effects may depend upon many factors, including the regimen of E2 utilized and interactions with the hypothalamic-pituitary-adrenal axis. Brain targets for E2's effects on anxiety and depression include the hippocampus and amygdala. Administration of E2, compared to vehicle, subcutaneously or to the hippocampus or amygdala of ovariectomized rats decreases anxiety and depressive behavior. Intracellular estrogen receptors (ERs) may be important for E2's anxiolytic and antidepressant-like effects. Administration of an ER antagonist to the hippocampus, but not amygdala, increases anxiety and depression behavior of naturally receptive female rats. Studies utilizing ER knockout mice or selective ER modulators suggest that ER-mediated effects of E2 on anxiety and depressive behavior may require ERbeta. In addition, the behavioral effects of E2 may involve membrane actions and/or changes in cell cycle processes involved in energy expenditure. Elucidating the mechanisms by which E2 affects anxiety and depression is important in order to enhance its therapeutic potential. It is particularly important to investigate the putative receptor mechanisms and brain targets for E2 to determine whether mood-enhancing effects of E2 can occur without deleterious proliferative effects in reproductive tissues.
Collapse
Affiliation(s)
- Alicia A Walf
- Department of Psychology, The University at Albany-SUNY, Albany, NY, USA
| | - Cheryl A Frye
- Department of Psychology, The University at Albany-SUNY, Albany, NY, USA
- Department of Biological Sciences, The University at Albany-SUNY, Albany, NY, USA
- Center for Neuroscience Research, The University at Albany-SUNY, Albany, NY, USA
- Center for Life Science Research, The University at Albany-SUNY, Albany, NY, USA
| |
Collapse
|
22
|
Dominguez-Salazar E, Shetty S, Rissman EF. Rapid neural Fos responses to oestradiol in oestrogen receptor alphabeta double knockout mice. J Neuroendocrinol 2006; 18:195-202. [PMID: 16454803 DOI: 10.1111/j.1365-2826.2005.01408.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The standard mode of action for oestradiol is via activation of nuclear oestrogen receptors (ERs), which initiate DNA transcription leading to protein formation. In the present study, we examined the rapid and potentially ER-independent action of oestradiol using Fos as a marker of neural activity. We assessed Fos immunoreactivity (ir) in brains of mice with functional versus nonfunctional ERs. Fos-ir was compared in brains of control mice that did and did not receive oestradiol treatment prior to sacrifice, and cell numbers in the preoptic area (POA), ventromedial nucleus of the hypothalamus (VMH), area 2 of cingulate cortex (CG2), granular layer of accessory olfactory bulb (Gr-AOB), olivary pretectal nucleus (OPT) and pyramidal layer of field CA3 of hippocampus (Py-CA3) were increased 90 min after oestradiol treatment. By contrast, in brains of double oestrogen receptor alphabeta knockout (ERalphabetaKO) female mice, no change in Fos-ir was noted after oestradiol treatment in the POA, VMH, Gr-AOB or Py-CA3, suggesting that these responses to oestradiol depend on ERalpha and/or ERbeta. However, Fos-ir was induced by oestradiol in the OPT and CG2 in ERalphabetaKO mice. These regions do not contain ERalpha-ir in control brains. In ERalphabetaKO brains as well, ERalpha-ir was absent, suggesting that the mutant ERalpha (E1) present in ERalphaKO brain is also absent in these regions. We speculate that oestradiol has rapid effects in the OPT and CG2 via a novel mechanism that does not require either classic oestrogen receptor.
Collapse
Affiliation(s)
- E Dominguez-Salazar
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
23
|
Auger CJ, Jessen HM, Auger AP. Microarray profiling of gene expression patterns in adult male rat brain following acute progesterone treatment. Brain Res 2005; 1067:58-66. [PMID: 16376865 DOI: 10.1016/j.brainres.2005.10.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Revised: 10/07/2005] [Accepted: 10/16/2005] [Indexed: 12/31/2022]
Abstract
Progesterone can influence various behaviors in adult male rats, however, little is known about which particular genes are regulated by progesterone in the male rat brain. Using focused microarray technology, we where able to define a subset of genes that are responsive to progesterone. Nylon membrane-based cDNA microarrays were used to profile gene expression patterns in the preoptic area/mediobasal hypothalamus (POA/MBH) of male rat brain 7 h following a single injection of progesterone. RNA was isolated from the brains of 6 male rats injected with progesterone and 6 male rats injected with sesame oil. Next, we hybridized the RNA from each animal to individual cDNA microarrays that contained more than 100 target genes, all of which are involved in cAMP and or calcium signaling pathways. Direct side-by-side comparison of all 12 arrays revealed differences in the expression patterns of 12 different genes. We confirmed the data gathered from the arrays on 4 different genes using Real-Time PCR. These data begin to outline the important role played by progesterone in mediating changes in gene expression within the male brain.
Collapse
Affiliation(s)
- Catherine J Auger
- Department of Zoology, Birge Hall, 430 Lincoln Drive, University of Wisconsin, Madison, WI 53706, USA.
| | | | | |
Collapse
|
24
|
Nakamura NH, Rosell DR, Akama KT, McEwen BS. Estrogen and ovariectomy regulate mRNA and protein of glutamic acid decarboxylases and cation-chloride cotransporters in the adult rat hippocampus. Neuroendocrinology 2005; 80:308-23. [PMID: 15677881 DOI: 10.1159/000083657] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2004] [Accepted: 10/28/2004] [Indexed: 12/22/2022]
Abstract
17beta-Estradiol spatiotemporally regulates the gamma-aminobutyric acid (GABAergic) tone in the adult hippocampus. However, the complex estrogenic effect on the GABAergic system is still unclear. In adult central nervous system (CNS) neurons, GABA can induce both inhibitory and excitatory actions, which are predominantly controlled by the cation-chloride cotransporters NKCC1 and KCC2. We therefore studied the estrogenic regulation of two glutamate decarboxylase (GAD) isoforms, GAD65 and GAD67, as well as NKCC1 and KCC2 in the adult female rat hippocampus by immunohistochemistry and in situ hybridization. First, we focused on the duration after ovariectomy (OVX) and its effects on GAD65 protein levels. The basal number of GAD65-immunoreactive cells decreased after long-term (10 days) OVX compared to short-term (3 days) OVX. We found that, only after long-term OVX but not after short-term OVX, estradiol increased the number of GAD65-immunoreactive cells in the CA1 pyramidal cell layer. Furthermore, estradiol did not alter the GAD65-immunoreactive cell population in any other CA1 subregion. Second, we therefore focused on long-term OVX and the estrogenic regulation of GAD and cation-chloride cotransporter mRNA levels. In the pyramidal cell layer, estradiol affected GAD65, GAD67 and NKCC1 mRNA levels, but not KCC2 mRNA levels. Both GAD65 and NKCC1 mRNA levels increased within 24 h after estradiol treatment, followed by a subsequent increase in GAD67 mRNA levels. These findings suggest that basal levels of estrogen might contribute to a balance between the excitatory and inhibitory synaptic transmission onto CA1 pyramidal cells by regulating perisomatic GAD and NKCC1 expression in the adult hippocampus.
Collapse
Affiliation(s)
- Nozomu H Nakamura
- Laboratory of Neuroendocrinology, Rockefeller University, 1230 York Avenue, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
25
|
Wu TW, Wang JM, Chen S, Brinton RD. 17β-estradiol induced Ca2+ influx via L-type calcium channels activates the Src/ERK/cyclic-AMP response element binding protein signal pathway and BCL-2 expression in rat hippocampal neurons: A potential initiation mechanism for estrogen-induced neuroprotection. Neuroscience 2005; 135:59-72. [PMID: 16084662 DOI: 10.1016/j.neuroscience.2004.12.027] [Citation(s) in RCA: 197] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2004] [Revised: 11/16/2004] [Accepted: 12/12/2004] [Indexed: 11/23/2022]
Abstract
Our group and others have demonstrated that 17beta-estradiol (E2) induces neurotrophic and neuroprotective responses in hippocampal and cortical neurons which are dependent upon the Src/extracellular signal-regulated kinase (ERK) signaling pathways. The purpose of this study was to determine the upstream mechanism(s) that initiates the signaling cascade leading to E2-inducible neuroprotection. We tested the hypothesis that E2 activates rapid Ca(2+) influx in hippocampal neurons, which would lead to activation of the Src/ERK signaling cascade and up-regulation of Bcl-2 protein expression. Using fura-2 ratiometric Ca(2+) imaging, we demonstrated that E2 induced a rapid rise of intracellular Ca(2+) concentration ([Ca(2+)](i)) within minutes of exposure which was blocked by an L-type Ca(2+) channel antagonist. Inhibition of L-type Ca(2+) channels resulted in a loss of E2 activation of the Src/ERK cascade, activation of cyclic-AMP response element binding protein (CREB) and subsequent increase in Bcl-2. Real-time intracellular Ca(2+) imaging combined with pERK immunofluorescence, demonstrated that E2 induced [Ca(2+)](i) was coincident with ERK activation in the same neuron. Small interfering RNA knockdown of CREB resulted in a loss of E2 activation of CREB and subsequent E2-induced increase of Bcl-2 expression. We further demonstrated the presence of specific membrane E2 binding sites in hippocampal neurons. Together, these data indicate that E2-induced Ca(2+) influx via the L-type Ca(2+) channel is required for E2 activation of the Src/ERK/CREB/Bcl-2 signaling. Implications of these data for understanding estrogen action in brain and use of estrogen therapy for prevention of neurodegenerative disease are discussed.
Collapse
Affiliation(s)
- T-W Wu
- Neuroscience Program, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089-9121, USA
| | | | | | | |
Collapse
|
26
|
Abstract
BACKGROUND Sex differences in the response threshold to painful stimuli and the higher number of chronic pain syndromes in women than in men have prompted a series of studies on lower animals and humans aimed at clarifying the role of gonadal hormones in pain. OBJECTIVE This article examines the morphologic and functional aspects of gonadal hormone systems and the relations between gonadal hormones and pain circuits, to identify areas deserving of increased attention in elucidating the endocrine mechanisms that contribute to abnormal pain states.
Collapse
|
27
|
Lâm TT, Leranth C. Gonadal hormones act extrinsic to the hippocampus to influence the density of hippocampal astroglial processes. Neuroscience 2003; 116:491-8. [PMID: 12559104 DOI: 10.1016/s0306-4522(02)00730-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The important effects of estrogen on the morphology of hippocampal neurons are well established. The mechanisms leading to such changes, nevertheless, have proved confusingly complex, since interactions between glia and neurons, as well as neuronal influences from other brain fields, are involved. This study addresses the possibility that estrogen-sensitive projections from the medial septum/diagonal band of Broca induce astroglial reactions. Estrogen- and cholesterol-filled (controls) cannulae were implanted into the medial septum/diagonal band of Broca of adult ovariectomized rats. Comparative semiquantitative immunohistochemical analysis on the density of the glial fibrillary acidic protein-containing processes and cells were performed on hippocampal slices of locally estrogen-treated and control animals. Rats that received estrogen-filled cannulae showed a lower density of glial processes in the hippocampal CA1 and CA3 subfields than animals of the control group. These effects could not be observed in the dentate gyrus. Cell counts revealed no significant difference in the number of glial fibrillary acidic protein-positive cells in any of the examined areas. Two major conclusions can be drawn from these results. First, the data show that estrogen, in fact, has an indirect influence on hippocampal cells through septo-hippocampal projections. Furthermore, estradiol can have an indirect negative effect on hippocampal astrocytes, causing a reduction in the density of their processes.
Collapse
Affiliation(s)
- T T Lâm
- Department of Obstetrics and Gynecology, Yale University, School of Medicine, New Haven, CT 06520-8063, USA
| | | |
Collapse
|
28
|
Rudick CN, Woolley CS. Selective estrogen receptor modulators regulate phasic activation of hippocampal CA1 pyramidal cells by estrogen. Endocrinology 2003; 144:179-87. [PMID: 12488344 DOI: 10.1210/en.2002-220581] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous studies demonstrated that estrogen induces two sequential waves of CA1 pyramidal cell activation, evidenced by induction of c-Fos at 2 and 24 h after a single estrogen treatment. The second wave of activation is paralleled by suppression of immunoreactivity for glutamic acid decarboxylase-65kD (GAD65) in CA1 and decreased synaptic inhibition of CA1 pyramidal cells. Here, we report that pretreatment with either of the selective estrogen receptor (ER) modulators, tamoxifen (T) or CI628, has no effect on the first wave of c-Fos expression at 2 h but completely blocks the second wave of c-Fos and the suppression of GAD65 at 24 h. Interestingly, T, given 4 h after estrogen, failed to block c-Fos expression or suppression of GAD65 at 24 h. Electrophysiological experiments showed that the T metabolite, 4OH-T, or CI628 can inhibit the so-called rapid estrogen effect, to potentiate excitatory postsynaptic currents (EPSCs) in CA1 pyramidal cells. Thus, estrogen seems to act within 4 h via classical ERs and/or a rapid estrogen effect, such as EPSC potentiation, to produce activation/disinhibition of pyramidal cells 24 h later. In contrast, the initial activation of pyramidal cells, at 2 h after estrogen, seems to involve neither classical ERs nor rapid potentiation of EPSCs.
Collapse
Affiliation(s)
- Charles N Rudick
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208, USA
| | | |
Collapse
|
29
|
Zheng H, Corkern MM, Crousillac SM, Patterson LM, Phifer CB, Berthoud HR. Neurochemical phenotype of hypothalamic neurons showing Fos expression 23 h after intracranial AgRP. Am J Physiol Regul Integr Comp Physiol 2002; 282:R1773-81. [PMID: 12010760 DOI: 10.1152/ajpregu.00019.2002] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Agouti-related protein (AgRP) is coexpressed with neuropeptide Y (NPY) in a population of neurons in the arcuate nucleus (ARC) of the hypothalamus and stimulates food intake for up to 7 days if injected intracerebroventricularly. The prolonged food intake stimulation does not seem to depend on continued competition at the melanocortin-4 receptor (MC4R), because the relatively specific MC4R agonist MTII regains its ability to suppress food intake 24 h after AgRP injection. Intracerebroventricular AgRP also stimulates c-Fos expression 24 h after injection in several brain areas, so the neurons exhibiting delayed Fos expression might be particularly important in feeding behavior. Thus we aimed to identify the neurochemical phenotype of some of these neurons in select hypothalamic areas, using double-label immunohistochemistry. AgRP-injected rats ingested significantly more chow (10.2 +/- 0.6 g) vs. saline controls (3.4 +/- 0.7 g) in the first 9 h (light phase) after injection. In the lateral hypothalamus (particularly the perifornical area) 23 h after injection, AgRP induced significantly more Fos vs. saline in orexin-A (OXA) neurons (25.6 +/- 4.9 vs. 4.8 +/- 3.1%), but not in melanin-concentrating hormone (MCH) or cocaine- and amphetamine-regulated transcript (CART) neurons. In the ARC, AgRP induced significantly more Fos in CART (40.6 +/- 5.9 vs. 13.4 +/- 1.8%) but not NPY neurons. In the paraventricular nucleus, there was no significant difference in Fos expression induced by AgRP vs. saline in oxytocin and CART neurons. We conclude that the long-lasting hyperphagia induced by AgRP is correlated with and possibly partially mediated by hyperactive OXA neurons in the lateral hypothalamus and CART neurons in the ARC, but not by NPY and MCH neurons. The substantial increase in light-phase food intake by AgRP supports a role for the arousing effects of OXA. Activation of CART neurons in the ARC (which likely coexpress proopiomelanocortin) could indicate attempts to activate counterregulatory decreases in food intake.
Collapse
Affiliation(s)
- Huiyuan Zheng
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana 70808, USA
| | | | | | | | | | | |
Collapse
|
30
|
Serova L, Rivkin M, Nakashima A, Sabban EL. Estradiol stimulates gene expression of norepinephrine biosynthetic enzymes in rat locus coeruleus. Neuroendocrinology 2002; 75:193-200. [PMID: 11914591 DOI: 10.1159/000048237] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gender-specific differences in susceptibility to a number of disorders related to catecholaminergic systems, including depression and hypertension, have been postulated to be mediated, at least in part, by estrogens. In this study, we examined if estrogens may regulate gene expression of norepinephrine biosynthetic enzymes. Administration of five injections of 15 or 40 microg/kg estradiol benzoate to ovariectomized (OVX) female rats elicited a dose-dependent elevation in mRNA levels of tyrosine hydroxylase (TH) in locus coeruleus, to as great as 3-fold over control. Dopamine beta-hydroxylase (DBH) mRNA levels were also similarly increased. To examine the mechanism, PC12 cells were cotransfected with luciferase reporter constructs under control of DBH or TH promoters [pDBH/Luc(-2,236/+21) or pTH/Luc(-272/+27 or -773/+27)] with an expression vector for estradiol receptor alpha. The cells were treated with 17beta-estradiol (E(2)) for 12-36 h. E(2) triggered a several fold increase in luciferase activity under control of the DBH promoter in a dose-dependent fashion. Omission of estrogen receptor alpha or addition of the estrogen receptor antagonist ICI 182,780 prevented the DBH promoter-driven increase in luciferase. When E(2) was given with 0.2 mM CPT-cAMP, reporter activity with pDBH/Luc(-2,236/+21) was increased greater than with either treatment alone. In contrast, addition of E(2) to cells transfected with pTH/Luc(-272/+27) elicited no change in basal luciferase activity nor in the response to 0.2 mM CPT-cAMP. These findings are the first to reveal that estrogen can stimulate DBH gene expression. Differing mechanisms may underlie the regulation of TH and DBH gene expression by estrogens.
Collapse
Affiliation(s)
- Lidia Serova
- Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
31
|
McEwen BS. Invited review: Estrogens effects on the brain: multiple sites and molecular mechanisms. J Appl Physiol (1985) 2001; 91:2785-801. [PMID: 11717247 DOI: 10.1152/jappl.2001.91.6.2785] [Citation(s) in RCA: 473] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Besides their well-established actions on reproductive functions, estrogens exert a variety of actions on many regions of the nervous system that influence higher cognitive function, pain mechanisms, fine motor skills, mood, and susceptibility to seizures; they also appear to have neuroprotective actions in relation to stroke damage and Alzheimer's disease. Estrogen actions are now recognized to occur via two different intracellular estrogen receptors, ER-alpha and ER-beta, that reside in the cell nuclei of some nerve cells, as well as by some less well-characterized mechanisms. In the hippocampus, such nerve cells are sparse in number and yet appear to exert a powerful influence on synapse formation by neurons that do not have high levels of nuclear estrogen receptors. However, we also find nonnuclear estrogen receptors outside of the cell nuclei in dendrites, presynaptic terminals, and glial cells, where estrogen receptors may couple to second messenger systems to regulate a variety of cellular events and signal to the nuclear via transcriptional regulators such as CREB. Sex differences exist in many of the actions of estrogens in the brain, and the process of sexual differentiation appears to affect many brain regions outside of the traditional brain areas involved in reproductive functions. Finally, the aging brain is responsive to actions of estrogens, which have neuroprotective effects both in vivo and in vitro. However, in an animal model, the actions of estrogens on the hippocampus appear to be somewhat attenuated with age. In the future, estrogen actions over puberty and in pregnancy and lactation should be further explored and should be studied in both the hypothalamus and the extrahypothalamic regions.
Collapse
Affiliation(s)
- B S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, 1230 York Ave., New York, NY 10021, USA.
| |
Collapse
|
32
|
Abstract
Dissociated cultured rat hippocampal pyramidal neurons respond to estradiol with a time-dependent, twofold increase in density of their dendritic spines. This effect is mediated by an estrogen receptor, probably of the alpha nuclear receptor type. In searching for the molecular mechanisms leading from the initial activation of the estrogen receptor to the final formation of new dendritic spines, we found that estradiol acts on GABAergic interneurons expressing the estrogen receptor by decreasing their inhibitory tone. In culture, this is assumed to cause a shift in the balance between excitation and inhibition toward enhanced excitation, overactivation of the pyramidal neurons, and subsequent formation of novel dendritic spines. The action of estradiol on spine formation is mediated by phosphorylation of cyclic AMP response element binding protein in the pyramidal neurons and is blocked when inhibition is enhanced by diazepam and when excitation is blocked by tetrodotoxin. Progesterone blocks the effect of estradiol on dendritic spines through its conversion to tetrahydroprogesterone, which enhances GABAergic inhibition. Subsequent to formation of novel dendritic spines, there is an increase in the density of glutamatergic receptors in the affected cells, an increase in the cellular calcium response to glutamate, and an increase in network synaptic activity among the cultured neurons.
Collapse
Affiliation(s)
- M Segal
- Neurobiology, The Weizmann Institute, Rehovot, 76100 Israel.
| | | |
Collapse
|
33
|
Solum DT, Handa RJ. Localization of estrogen receptor alpha (ER alpha) in pyramidal neurons of the developing rat hippocampus. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2001; 128:165-75. [PMID: 11412902 DOI: 10.1016/s0165-3806(01)00171-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
During development, estrogen has a variety of effects on morphological, biochemical and electrophysiological properties of hippocampal neurons. Correspondingly, estrogen receptor (ER) binding and mRNA increase transiently in the developing hippocampus. In this study, we used immunocytochemistry to determine the localization of the ER alpha subtype in the developing rat hippocampus. Nuclear staining was present in pyramidal cells and some interneurons of the CA1 and CA3 regions of the developing rat hippocampus. Little or no immunoreactivity was observed in postnatal day (P)0 animals (day of birth=P0), however, beginning on P4, ER alpha-immunoreactivity (ER alpha-ir) was visible and reached maximal levels by P10. These levels subsequently declined to low levels so that by P15, levels approximated those of adult females. Western blot analysis confirmed that this antibody recognized a 67 kDa protein, characteristic of the full-length ER alpha protein, in the hippocampus and pituitary. Furthermore, most of the ER alpha-immunopositive cells in the hippocampus were located in the pyramidal cell layer, and did not co-localize appreciably with gamma-aminobutyric acid (GABA) at any age examined. We conclude, based on the immunocytochemical localization of ER alpha, that the effects of estrogen on biochemistry and morphology of the developing hippocampus may be direct through the ER alpha subtype in hippocampal pyramidal cells.
Collapse
Affiliation(s)
- D T Solum
- Department of Cell Biology, Neurobiology and Anatomy, Loyola University Chicago, Maywood, IL 60301, USA
| | | |
Collapse
|
34
|
McEwen B, Akama K, Alves S, Brake WG, Bulloch K, Lee S, Li C, Yuen G, Milner TA. Tracking the estrogen receptor in neurons: implications for estrogen-induced synapse formation. Proc Natl Acad Sci U S A 2001; 98:7093-100. [PMID: 11416193 PMCID: PMC34628 DOI: 10.1073/pnas.121146898] [Citation(s) in RCA: 249] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Estrogens (E) and progestins regulate synaptogenesis in the CA1 region of the dorsal hippocampus during the estrous cycle of the female rat, and the functional consequences include changes in neurotransmission and memory. Synapse formation has been demonstrated by using the Golgi technique, dye filling of cells, electron microscopy, and radioimmunocytochemistry. N-methyl-d-aspartate (NMDA) receptor activation is required, and inhibitory interneurons play a pivotal role as they express nuclear estrogen receptor alpha (ERalpha) and show E-induced decreases of GABAergic activity. Although global decreases in inhibitory tone may be important, a more local role for E in CA1 neurons seems likely. The rat hippocampus expresses both ERalpha and ERbeta mRNA. At the light microscopic level, autoradiography shows cell nuclear [3H]estrogen and [125I]estrogen uptake according to a distribution that primarily reflects the localization of ERalpha-immunoreactive interneurons in the hippocampus. However, recent ultrastructural studies have revealed extranuclear ERalpha immunoreactivity (IR) within select dendritic spines on hippocampal principal cells, axon terminals, and glial processes, localizations that would not be detectable by using standard light microscopic methods. Based on recent studies showing that both types of ER are expressed in a form that activates second messenger systems, these findings support a testable model in which local, non-genomic regulation by estrogen participates along with genomic actions of estrogens in the regulation of synapse formation.
Collapse
Affiliation(s)
- B McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Auger AP, Hexter DP, McCarthy MM. Sex difference in the phosphorylation of cAMP response element binding protein (CREB) in neonatal rat brain. Brain Res 2001; 890:110-7. [PMID: 11164773 DOI: 10.1016/s0006-8993(00)03151-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
On the day of birth, a surge in testicular testosterone release in male rats is critical for sexual differentiation of the brain. Steroid hormones function by binding to intracellular steroid receptors and altering gene expression; however, little is known about the signal transduction pathways altered as a consequence of steroid hormone action in developing brain. We investigated whether the increase in testosterone at birth alters the phosphorylation of CREB, a major signal transduction protein. Adjacent brain sections from male and female pups were immunocytochemically stained for serine(133) phosphorylated CREB (pCREB) or total CREB on the day of birth. Males had more pCREB-immunoreactive positive cells than females in the medial preoptic area, ventromedial nucleus of the hypothalamus, the arcuate nucleus, and the CA1 region of the hippocampus, but not in two thalamic nuclei. There was no sex difference in total CREB immunoreactive cell number. To determine if the sex difference in pCREB persisted 24 h after birth and whether the difference was due to testosterone, newborn female pups were injected with 100 microg of testosterone propionate, and male and control female pups were injected with vehicle. Twenty-four hours later, adjacent brain sections were immunocytochemically stained for either pCREB or CREB. We found that males and testosterone-treated females had more pCREB in the ventromedial nucleus of the hypothalamus contrasted to control females. There were no group differences in pCREB or CREB in any other area examined. These results indicate that some of the effects of testosterone in developing brain occur via pathways associated with the phosphorylation of CREB.
Collapse
Affiliation(s)
- A P Auger
- Department of Physiology, School of Medicine, University of Maryland-Baltimore, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|