51
|
Hucho TB, Dina OA, Kuhn J, Levine JD. Estrogen controls PKCepsilon-dependent mechanical hyperalgesia through direct action on nociceptive neurons. Eur J Neurosci 2006; 24:527-34. [PMID: 16836642 DOI: 10.1111/j.1460-9568.2006.04913.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protein kinase C epsilon (PKCepsilon) is an important intracellular signaling molecule in primary afferent nociceptors, implicated in acute and chronic inflammatory as well as neuropathic pain. In behavioral experiments inflammatory mediators produce PKCepsilon-dependent hyperalgesia only in male rats. The mechanism underlying this sexual dimorphism is unknown. We show that the hormone environment of female rats changes the nociceptive signaling in the peripheral sensory neuron. This change is maintained in culture also in the absence of a gender-simulating environment. Stimulation of beta(2)-adrenergic receptors (beta(2)-AR) leads to PKCepsilon activation in cultured dorsal root ganglia (DRG) neurons derived from male but not from female rats. Addition of estrogen to male DRG neurons produces a switch to the female phenotype, namely abrogation of beta(2)-AR-initiated activation of PKCepsilon. Estrogen interferes downstream of the beta(2)-AR with the signaling pathway leading from exchange protein activated by cAMP (Epac) to PKCepsilon. The interfering action is fast indicating a transcriptional-independent mechanism. Estrogen has a dual effect on PKCepsilon. If applied before beta(2)-AR or Epac stimulation, estrogen abrogates the activation of PKCepsilon. In contrast, estrogen applied alone leads to a brief translocation of PKCepsilon. Also in vivo the activity of estrogen depends on the stimulation context. In male rats, intradermal injection of an Epac activator or estrogen alone induces mechanical hyperalgesia through a PKCepsilon-dependent mechanism. In contrast, injection of estrogen preceding the activation of Epac completely abrogates the Epac-induced mechanical hyperalgesia. Our results suggest that gender differences in nociception do not reflect the use of generally different mechanisms. Instead, a common set of signaling pathways can be modulated by hormones.
Collapse
Affiliation(s)
- Tim B Hucho
- NIH Pain Center, UCSF, University of California, San Francisco, 521 Parnassus Avenue, PO-Box 0440, 94143, USA.
| | | | | | | |
Collapse
|
52
|
Galanopoulou AS. Sex- and cell-type-specific patterns of GABAAreceptor and estradiol-mediated signaling in the immature rat substantia nigra. Eur J Neurosci 2006; 23:2423-30. [PMID: 16706849 DOI: 10.1111/j.1460-9568.2006.04778.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The substantia nigra pars reticulata (SNR) is involved in movement and seizure control. In male but not female postnatal day 15 (PN15) rats, GABAA receptor agonists depolarize the SNR neurons and increase the expression of the calcium-regulated gene KCC2 (potassium/chloride cotransporter). Moreover, in PN15 rat SNR, 7beta-estradiol down-regulates KCC2 expression only in the presence of depolarizing GABAA receptor responses. The hypothesis tested here was that GABAA receptors and estradiol also regulate the expression of the phosphorylated form of the transcription factor cAMP responsive element binding protein (phosphoCREB), in PN15 rat SNR and substantia nigra pars compacta (SNC). Rats were injected with muscimol or 17beta-estradiol or their vehicles, and killed 1 h later. Sections were stained with an antibody specific for phosphoCREB alone or counterstained with either tyrosine hydroxylase (TH)- or parvalbumin (PRV)-specific antibodies. Muscimol increased phosphoCREB-ir in male but not in female SN neurons. Using gramicidin perforated patch clamp of PN14-15 SNC neuron, it was shown that muscimol bath application depolarized male SNC neurons but did not significantly alter membrane potential in females. In males, 17beta-estradiol decreased phosphoCREB expression in all studied cell types. In females, 17beta-estradiol did not influence phosphoCREB expression in PRV-ir SNR cells, but increased it in the dopaminergic SN neurons. These data suggest that GABAA receptor activation and estradiol promote the sexual differentiation of the SN in a cell-type-specific manner, by influencing calcium-regulated gene transcription, and therefore promoting the acquisition of sex-specific roles of the SN in movement and seizure control.
Collapse
Affiliation(s)
- Aristea S Galanopoulou
- Department of Neurology and Einstein/Montefiore Comprehensive Epilepsy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Kennedy Center Rm 311, Bronx, NY 10461, USA.
| |
Collapse
|
53
|
Bryant DN, Sheldahl LC, Marriott LK, Shapiro RA, Dorsa DM. Multiple pathways transmit neuroprotective effects of gonadal steroids. Endocrine 2006; 29:199-207. [PMID: 16785596 DOI: 10.1385/endo:29:2:199] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 11/30/1999] [Accepted: 10/25/2005] [Indexed: 12/27/2022]
Abstract
Numerous preclinical studies suggest that gonadal steroids, particularly estrogen, may be neuroprotective against insult or disease progression. This paper reviews the mechanisms contributing to estrogen-mediated neuroprotection. Rapid signaling pathways, such as MAPK, PI3K, Akt, and PKC, are required for estrogen's ability to provide neuroprotection. These rapid signaling pathways converge on genomic pathways to modulate transcription of E2-responsive genes via ERE-dependent and ERE-independent mechanisms. It is clear that both rapid signaling and transcription are important for estrogen's neuroprotective effects. A mechanistic understanding of estrogen-mediated neuroprotection is crucial for the development of therapeutic interventions that enhance quality of life without deleterious side effects.
Collapse
Affiliation(s)
- Damani N Bryant
- Department of Physiology and Pharmacology (L334), Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
54
|
Thomas W, Coen N, Faherty S, Flatharta CO, Harvey BJ. Estrogen induces phospholipase A2 activation through ERK1/2 to mobilize intracellular calcium in MCF-7 cells. Steroids 2006; 71:256-65. [PMID: 16375935 DOI: 10.1016/j.steroids.2005.10.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Revised: 10/05/2005] [Accepted: 10/31/2005] [Indexed: 12/29/2022]
Abstract
The principal secreted estrogen, 17beta-estradiol rapidly activates signaling cascades that regulate important physiological processes including ion transport across membranes, cytosolic pH and cell proliferation. These effects have been extensively studied in the MCF-7 estrogen-responsive human breast carcinoma cell line. Here, we demonstrate that a physiological concentration of 17beta-estradiol caused a rapid, synchronous and transient increase in intracellular calcium concentration in a confluent monolayer of MCF-7 cells 2-3 min after treatment. This response was abolished when cells were pre-incubated with the phospholipase A(2) (PLA(2)) inhibitor quinacrine or with the cyclooxygenase inhibitor indomethacin. The translocation of GFP-cPLA(2)alpha to perinuclear membranes occurred 1-2 min after 17beta-estradiol treatment; this translocation was concurrent with the transient phosphorylation of cPLA(2)alpha at serine residue 505. The phosphorylation and translocation of cPLA(2) were sensitive to inhibition of the extracellular signal regulated kinase (ERK) signaling cascade and occurred simultaneously with a transient activation of ERK. The phosphorylation of cPLA(2) could be stimulated by membrane impermeable 17beta-estradiol conjugated to bovine serum albumen and was blocked by an antagonist of the classical estrogen receptor. Here we show, for the first time, that PLA(2) and the eicosanoid biosynthetic pathway are involved in the 17beta-estradiol induced rapid calcium responses of breast cancer cells.
Collapse
Affiliation(s)
- Warren Thomas
- Charitable Infirmary Trust Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Beaumont Hospital, P.O. Box 9063, Dublin 9, Ireland.
| | | | | | | | | |
Collapse
|
55
|
Cornil CA, Taziaux M, Baillien M, Ball GF, Balthazart J. Rapid effects of aromatase inhibition on male reproductive behaviors in Japanese quail. Horm Behav 2006; 49:45-67. [PMID: 15963995 PMCID: PMC3515763 DOI: 10.1016/j.yhbeh.2005.05.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2004] [Revised: 04/27/2005] [Accepted: 05/03/2005] [Indexed: 10/25/2022]
Abstract
Non-genomic effects of steroid hormones on cell physiology have been reported in the brain. However, relatively little is known about the behavioral significance of these actions. Male sexual behavior is activated by testosterone partly through its conversion to estradiol via the enzyme aromatase in the preoptic area (POA). Brain aromatase activity (AA) changes rapidly which might in turn be important for the rapid regulation of behavior. Here, acute effects of Vorozole, an aromatase inhibitor, injected IP at different doses and times before testing (between 15 and 60 min), were assessed on male sexual behavior in quail. To limit the risk of committing both types of statistical errors (I and II), data of all experiments were entered into a meta-analysis. Vorozole significantly inhibited mount attempts (P < 0.05, size effect [g] = 0.527) and increased the latency to first copulation (P < 0.05, g = 0.251). The treatment had no effect on the other measures of copulatory behavior. Vorozole also inhibited appetitive sexual behavior measured by the social proximity response (P < 0.05, g = 0.534) or rhythmic cloacal sphincter movements (P < 0.001, g = 0.408). Behavioral inhibitions always reached a maximum at 30 min. Another aromatase inhibitor, androstatrienedione, induced a similar rapid inhibition of sphincter movements. Radioenzyme assays demonstrated that within 30 min Vorozole had reached the POA and completely blocked AA measured in homogenates. When added to the extracellular milieu, Vorozole also blocked within 5 min the AA in POA explants maintained in vitro. Together, these data demonstrate that aromatase inhibition rapidly decreases both consummatory and appetitive aspects of male sexual behavior.
Collapse
Affiliation(s)
- Charlotte A Cornil
- Center for Cellular and Molecular Neurobiology, Research Group in Behavioral Neuroendocrinology, University of Liège, Belgium.
| | | | | | | | | |
Collapse
|
56
|
Garcia-Segura LM, Sanz A, Mendez P. Cross-talk between IGF-I and estradiol in the brain: focus on neuroprotection. Neuroendocrinology 2006; 84:275-9. [PMID: 17124377 DOI: 10.1159/000097485] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Accepted: 10/17/2006] [Indexed: 12/27/2022]
Abstract
The actions of estradiol in the brain involve the interaction with growth factors, such as insulin-like growth factor-I (IGF-I). Many cells in the brain coexpress receptors for estradiol (ERs) and IGF-I (IGF-IR) and both factors interact to regulate neural function. Several studies have shown that there is an interaction of IGF-IR and ERs in neuroprotection. Neuroprotective effects of estradiol are blocked by the inhibition of IGF-IR signaling, while the neuroprotective effects of IGF-I are blocked by the inhibition of ER signaling. These findings suggest that the neuroprotective actions of estradiol and IGF-I after brain injury depend on the coactivation of both ERs and IGF-IR in neural cells. The relationship of ERalpha with IGF-IR through the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta (PI3K/Akt/GSK3) signaling pathway may represent the point of convergence used by estradiol and IGF-I to cooperatively promote neuroprotection. Administration of estradiol to ovariectomized rats results in the association of ERalpha with IGF-IR and with components of the PI3K/Akt/GSK3 signaling pathway and in the regulation of the activity of Akt and GSK3 in the brain. Conversely, IGF-I regulates ERalpha transcriptional activity in neuroblastoma cells and the PI3K/Akt/GSK3 signaling pathway is involved in this effect.
Collapse
Affiliation(s)
- Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| | | | | |
Collapse
|
57
|
Cordey M, Pike CJ. Conventional protein kinase C isoforms mediate neuroprotection induced by phorbol ester and estrogen. J Neurochem 2005; 96:204-17. [PMID: 16336227 DOI: 10.1111/j.1471-4159.2005.03545.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Rapid signal transduction pathways play a prominent role in mediating neuroprotective actions of estrogen in the CNS. We have previously shown that estrogen-induced neuroprotection of primary cerebrocortical neurons from beta-amyloid peptide (Abeta) toxicity depends on activation of protein kinase C (PKC). PKC activation with phorbol-12-myristate-13-acetate (PMA) also provides neuroprotection in this paradigm. Because the PKC family includes several isoforms that have opposing roles in regulating cell survival, we sought to identify which PKC isoforms contribute to neuroprotection induced by PMA and estrogen. We detected protein expression of multiple PKC isoforms in primary neuron cultures, including conventional (alpha, betaI, betaII), novel (delta, epsilon, theta) and atypical (zeta, iota/lambda) PKC. Using a panel of isoform-specific peptide inhibitors and activators, we find that novel and atypical PKC isoforms do not participate in the mechanism of either PMA or estrogen neuroprotection. In contrast, a selective peptide activator of conventional PKC isoforms provides dose-dependent neuroprotection against Abeta toxicity. In addition, peptide inhibitors of conventional, betaI, or betaII PKC isoforms significantly reduce protection afforded by PMA or 17beta-estradiol. Taken together, these data provide evidence that conventional PKC isoforms mediate phorbol ester and estrogen neuroprotection of cultured neurons challenged by Abeta toxicity.
Collapse
Affiliation(s)
- Myriam Cordey
- Neuroscience Graduate Program and Andrus Gerontology Center, University of Southern California, Los Angeles, California 90089-0191, USA
| | | |
Collapse
|
58
|
Pretorius E, Bornman MS. Calcium-mediated aponecrosis plays a central role in the pathogenesis of estrogenic chemical-induced neurotoxicity. Med Hypotheses 2005; 65:893-904. [PMID: 16051444 DOI: 10.1016/j.mehy.2005.03.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Accepted: 03/10/2005] [Indexed: 12/28/2022]
Abstract
Estrogen is traditionally associated with females but is also present in males, and influences aspects of brain chemistry and brain morphology in males, females and also during prenatal development. Humans as well as animals are additionally exposed to environmental products that mimic estrogen activity, also known as endocrine disrupters (EDCs). This hypothesis article investigates the role of estrogen (and also EDCs) in the brain and how it influences the Ca2+ pathway. Ca2+ and its movement in and out of the cell is an extremely important ion controlling normal cell physiology. Any dysfunction in the movement from outside to inside the cell or between organelles may have fundamentally negative effects and the disturbance may even lead to apoptosis and/or necrosis. Therefore we consider whether estrogen and EDCs may alter the Ca2+ physiology and whether these changes may be one of the main causes of interference in physiology that is seen when humans and animals are exposed to EDCs. We come to the conclusion that on a molecular level Ca2+ and Ca2+ fluxes ([Ca2+]i, endocrine disrupting chemicals, redox modulation, mitochondria and cytochrome c followed by apoptosis, necrosis or most likely aponecrosis may contribute to chemical-mediated developmental toxicity. Similarly, we hypothesize that calcium-mediated aponecrosis do not only play a central role in the pathophysiology of estrogenic chemical-induced neurotoxicity, but can contribute to chemical-mediated developmental toxicity in general, thereby affecting almost all cells and organs of the living organism.
Collapse
Affiliation(s)
- E Pretorius
- Department of Anatomy, School of Health Sciences, Medical Faculty of the University of Pretoria, P.O. Box 2034, BMW Building, Pretoria 0001, South Africa.
| | | |
Collapse
|
59
|
Viso-León MC, Ripoll C, Nadal A. Oestradiol rapidly inhibits Ca2+ signals in ciliary neurons through classical oestrogen receptors in cytoplasm. Pflugers Arch 2005; 449:33-41. [PMID: 15258764 DOI: 10.1007/s00424-004-1308-z] [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] [Indexed: 11/26/2022]
Abstract
Oestrogen plays a key role in a great variety of actions in the nervous system, either through classical or alternative pathways. The classical pathways are initiated after oestrogen binding to the oestrogen receptors ERalpha or ERbeta, which translocate from the cytoplasm to the nucleus and act there as transcription factors. Alternative pathways are initiated at the plasma membrane and cytoplasm, via binding to classical or non-classical ERs. Using isolated ciliary ganglion neurons from the chick embryo and Ca2+ imaging, we demonstrated that a 10-min exposure to 17beta-oestradiol reduces Ca2+ influx through the plasma membrane. This effect was not reproduced by oestradiol conjugated to bovine serum albumin, which does not cross the plasma membrane, indicating that 17beta-oestradiol was acting intracellularly. ERalpha was detected in the cytoplasm by immunostaining and its involvement in the regulation of Ca2+ influx by ICI182,780 inhibition. The phosphatidylinositol-3 kinase (Pi3-kinase) inhibitor wortmannin and the nitric oxide synthase (NOS) inhibitor Nomega-nitro-L-arginine methyl ester (L-NAME) both blocked the oestradiol effect. The oestradiol effect was reproduced by 8Br-cGMP and abolished in the presence of the cGMP-dependent protein kinase (PKG) inhibitor KT5823. Our study indicates that 17beta-oestradiol can regulate Ca2+ influx via PI3-kinase, NOS and PKG after activation of cytoplasmic ER.
Collapse
Affiliation(s)
- M Carmen Viso-León
- Institut de Bioenginyeria, Universitat Miguel Hernández d'Elx, Campus de Sant Joan, Carretera Alacant-Valéncia Km 87, 03550 Sant Joan d'Alacant, Spain
| | | | | |
Collapse
|
60
|
Pawlak J, Karolczak M, Krust A, Chambon P, Beyer C. Estrogen receptor-alpha is associated with the plasma membrane of astrocytes and coupled to the MAP/Src-kinase pathway. Glia 2005; 50:270-5. [PMID: 15712205 DOI: 10.1002/glia.20162] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Estrogens influence CNS development and a broad spectrum of neural functions. Several lines of evidence also suggest a neuroprotective role for estrogen. Different modes of estrogen action have been described at the cellular level involving classical nuclear estrogen receptor (ER)-dependent and nonclassical membrane ER-mediated rapid signaling. We have previously shown that nonclassical estrogen signaling is implicated in the control of dopamine cell function and protection. Since nonclassical interactions between estrogens and glia may contribute to these effects, our aim was to demonstrate the presence of membrane-associated ERs and their putative coupling to intracellular signaling pathways in astrocytes. Confocal image analysis and fluorescence-activated cell sorting (FACS) studies indicated the attachment of ER-alpha but not ER-beta to the plasma membrane of astrocytes. ERs were located in the cell soma region and glial processes. FACS analysis revealed that only a subpopulation of midbrain astrocytes possesses membrane ER-alpha. In FACS studies on ER-alpha knockout astrocytes, only a few membrane ER-positive cells were detected. The activation of membrane ERs appears to be coupled to the MAP-kinase/Src signaling pathway as shown by Western blotting. In conclusion, our data provide good evidence that nonclassical estrogen action in astrocytes is mediated by membrane ER-alpha. The physiological consequence of this phenomenon is not yet understood, but it might have a pivotal role in estrogen-mediated protective effects on midbrain dopamine neurons.
Collapse
Affiliation(s)
- Justyna Pawlak
- Anatomisches Institut, Universität Tübingen, D-72047 Tübingen, Germany
| | | | | | | | | |
Collapse
|
61
|
Pawlak J, Beyer C. Developmental expression of MNAR mRNA in the mouse brain. Cell Tissue Res 2005; 320:545-9. [PMID: 15846512 DOI: 10.1007/s00441-005-1090-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2004] [Accepted: 01/21/2005] [Indexed: 10/25/2022]
Abstract
During the development of the central nervous system, estrogen influences cellular differentiation and determines the functional connectivity of distinct neural networks. Estrogens generally act through nuclear estrogen receptors (ERs). Recent research has additionally revealed rapid estrogen effects requiring the binding of estrogen to membrane/cytoplasmic ERs and the activation of intracellular signaling systems such as the Src/MAPK cascade. The scaffold protein MNAR/PELP1 appears to be the designated functional mediator of such non-genomic estrogen effects between non-nuclear ERs and Src/MAPKs. In this study, we demonstrate the expression and differential regulation of MNAR mRNA in the developing male and female mouse brain by quantitative polymerase chain reaction. In the midbrain and hypothalamus, a gradual decline in MNAR mRNA levels has been observed prenatally with the highest values at embryonic day 15 and lowest at postnatal day 15. In the cortex, mRNA levels do not fluctuate until postnatal day 7 but decrease thereafter. No differences in MNAR expression between sexes have been detected. Analysis of neuronal and astroglia-enriched cell cultures has revealed the presence of MNAR in both cell types.
Collapse
Affiliation(s)
- Justyna Pawlak
- Anatomisches Institut, Universität Tübingen, Osterbergstrasse 3, 72074, Tübingen, Germany
| | | |
Collapse
|
62
|
Bulayeva NN, Wozniak AL, Lash LL, Watson CS. Mechanisms of membrane estrogen receptor-alpha-mediated rapid stimulation of Ca2+ levels and prolactin release in a pituitary cell line. Am J Physiol Endocrinol Metab 2005; 288:E388-97. [PMID: 15494610 DOI: 10.1152/ajpendo.00349.2004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of membrane estrogen receptor-alpha (mERalpha) in rapid nongenomic responses to 17beta-estradiol (E(2)) was tested in sublines of GH3/B6 rat prolactinoma cells selected for high (GH3/B6/F10) and low (GH3/B6/D9) mERalpha expression. E(2) elicited rapid, concentration-dependent intracellular Ca(2+) concentration ([Ca(2+)](i)) increases in the F10 subline. Lack of inhibition by thapsigargin depletion of intracellular Ca(2+) pools, together with abrogation of the response in Ca(2+)-free medium, suggested an extracellular source of Ca(2+) for this response. The participation of voltage-dependent channels in the E(2)-induced [Ca(2+)](i) increase was confirmed by the specific L-type Ca(2+) channel inhibitor nifedipine. For comparison, the D9 mERalpha-depleted subline was insensitive to steroid action via this signaling mechanism. [Ca(2+)](i) elevation was correlated with prolactin (PRL) release in the F10 cell line in as little as 3 min. E(2) caused a much higher PRL release than KCl treatment (which caused maximal Ca(2+) elevation), suggesting that secretion was also controlled by additional mechanisms. Participation of mERalpha in these effects was confirmed by the ability of E(2)-peroxidase (a cell-impermeable analog of E(2)) to cause these responses, blockage of the responses with the ER antagonist ICI 182 780, and the inability of the E(2) stereoisomer 17alpha-E(2) to elicit a response. Thus rapid exocytosis of PRL is regulated in these cells by mERalpha signaling to specific Ca(2+) channels utilizing extracellular Ca(2+) sources and additional signaling mechanisms.
Collapse
Affiliation(s)
- Nataliya N Bulayeva
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0645, USA
| | | | | | | |
Collapse
|
63
|
Zhang S, Huang Y, Zhu YC, Yao T. Estrogen stimulates release of secreted amyloid precursor protein from primary rat cortical neurons via protein kinase C pathway. Acta Pharmacol Sin 2005; 26:171-6. [PMID: 15663894 DOI: 10.1111/j.1745-7254.2005.00538.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM To investigate the mechanism of the action of estrogen, which stimulates the release of secreted amyloid precursor protein alpha (sAPP(alpha)) and decreases the generation of amyloid-beta protein (A(beta)), a dominant component in senile plaques in the brains of Alzheimer's disease patients. METHODS Experiments were carried out in primary rat cortical neurons, and Western blot was used to detect sAPP(alpha) in a culture medium and the total amount of cellular amyloid precursor protein (APP) in neurons. RESULTS 17beta-Estradiol (but not 17alpha-estradiol) and beta-estradiol 6-(O-carboxymethyl) oxime: BSA increased the secretion of sAPP(alpha) and this effect was blocked by protein kinase C (PKC) inhibitor calphostin C, but not by the classical estrogen receptor antagonist ICI 182,780. Meanwhile, 17beta-estradiol did not alter the synthesis of cellular APP. CONCLUSION The effect of 17beta-estradiol on sAPP(alpha) secretion is likely mediated through the membrane binding sites, and needs molecular configuration specificity of the ligand. Furthermore, the action of the PKC-dependent pathway might be involved in estrogen-induced sAPP(alpha) secretion.
Collapse
Affiliation(s)
- Sun Zhang
- Department of Physiology and Pathophysiology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | | | | | | |
Collapse
|
64
|
D'Astous M, Morissette M, Callier S, Di Paolo T. Regulation of striatal preproenkephalin mRNA levels in MPTP-lesioned mice treated with estradiol. J Neurosci Res 2005; 80:138-44. [PMID: 15723346 DOI: 10.1002/jnr.20412] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We reported previously the protective effect of 17beta-estradiol (17beta-E(2)) on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopamine (DA) depletion. This protection was stereospecific, because 17beta-E(2) showed activity but 17alpha-estradiol (17alpha-E(2)) did not. The mechanisms by which estradiol exerts its beneficial effects, however, remain unknown. We investigated a possible implication of enkephalins (ENK) in neuroprotective activity of 17beta-E(2). Protection against MPTP-induced DA depletion was obtained with 17beta-E(2) but not 17alpha-E(2). MPTP lesion increased striatal preproenkephalin (PPE) mRNA levels and they remained elevated in 17alpha-E(2)-treated MPTP mice whereas 17beta-E(2) treatment decreased these levels to control values. This is the first report of estradiol modulation of striatal PPE mRNA in mice. Negative and significant correlations between DA levels, vesicular monoamine transporter (VMAT(2)) density, and PPE mRNA were observed in the striatum of lesioned animals. This effect of 17beta-E(2) on PPE mRNA after a lesion could be one of many mechanisms by which this steroid exerts its neuroprotective activity.
Collapse
Affiliation(s)
- Myreille D'Astous
- Molecular Endocrinology and Oncology Research Center, Laval University Medical Center, CHUL, Quebec City, Quebec, Canada
| | | | | | | |
Collapse
|
65
|
Zhao X, MacBride MM, Peterson BR, Pfaff DW, Vasudevan N. Calcium flux in neuroblastoma cells is a coupling mechanism between non-genomic and genomic modes of estrogens. Neuroendocrinology 2005; 81:174-82. [PMID: 16020926 DOI: 10.1159/000087000] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2004] [Accepted: 03/19/2005] [Indexed: 11/19/2022]
Abstract
Estrogens have been demonstrated to rapidly modulate calcium levels in a variety of cell types. However, the significance of estrogen-mediated calcium flux in neuronal cells is largely unknown. The relative importance of intra- and extracellular sources of calcium in estrogenic effects on neurons is also not well understood. Previously, we have demonstrated that membrane-limited estrogens, such as E-BSA given before an administration of a 2-hour pulse of 17beta-estradiol (E2), can potentiate the transcription mediated by E2 from a consensus estrogen response element (ERE)-driven reporter gene. Inhibitors to signal transduction cascades given along with E-BSA or E2 demonstrated that calcium flux is important for E-BSA-mediated potentiation of transcription in a transiently transfected neuroblastoma cell line. In this report, we have used inhibitors to different voltage-gated calcium channels (VGCCs) and to intracellular store receptors along with E-BSA in the first pulse or with E2 in the second pulse to investigate the relative importance of these channels to estrogen-mediated transcription. Neither L- nor P-type VGCCs seem to play a role in estrogen action in these cells; while N-type VGCCs are important in both the non-genomic and genomic modes of estrogen action. Specific inhibitors also showed that the ryanodine receptor and the inositol trisphosphate receptor are important to E-BSA-mediated transcriptional potentiation. This report provides evidence that while intracellular stores of calcium are required to couple non-genomic actions of estrogen initiated at the membrane to transcription in the nucleus, extracellular sources of calcium are also important in both non-genomic and genomic actions of estrogens.
Collapse
Affiliation(s)
- Xing Zhao
- Department of Biology, Pennsylvania State University, University Park, PA 16802 , USA
| | | | | | | | | |
Collapse
|
66
|
Koldzic-Zivanovic N, Seitz PK, Watson CS, Cunningham KA, Thomas ML. Intracellular signaling involved in estrogen regulation of serotonin reuptake. Mol Cell Endocrinol 2004; 226:33-42. [PMID: 15489003 DOI: 10.1016/j.mce.2004.07.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2004] [Revised: 07/06/2004] [Accepted: 07/08/2004] [Indexed: 11/15/2022]
Abstract
17beta-estradiol (E2) regulates neuronal activity via genomic and rapid, non-genomic mechanisms. The rat serotonergic neuronal cell line (RN46A) was used to investigate the rapid effects of E2 on serotonin (5-HT) reuptake and on potential intracellular signaling pathways. RN46A cells express the serotonin transporter (SERT) and estrogen receptor (ER)beta, but not ERalpha. Fifteen minute E2 treatment (10(-9)M) decreased 5-HT uptake. Intracellular cAMP levels were not increased by 15 min E2 treatment; however, E2 caused an increase in intracellular Ca2+ levels, with a maximum response within the first minute. The response was E2 specific, since other steroids (17alpha-estradiol, testosterone, and progesterone) had no effect. The ER antagonist ICI 182,780 blocked the rapid E2 effects on intracellular Ca2+ levels as did the selective ER modulator tamoxifen. In summary, changes in intracellular Ca2+ levels caused by E2 and mediated through ERbeta may be responsible for observed rapid effects of E2 on SERT activity.
Collapse
Affiliation(s)
- Nina Koldzic-Zivanovic
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-1031, USA
| | | | | | | | | |
Collapse
|
67
|
Ba F, Pang PKT, Davidge ST, Benishin CG. The neuroprotective effects of estrogen in SK-N-SH neuroblastoma cell cultures. Neurochem Int 2004; 44:401-11. [PMID: 14687605 DOI: 10.1016/j.neuint.2003.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Estrogen has been considered to be a neuroprotectant and a neuromodulator in many neuronal cell lines and tissue preparations. The protective effects of estrogen may be mediated through classical estrogen receptors (ERs), or may be due to its anti-oxidant properties which are independent of receptors. The current studies show that 17beta-estradiol (E2) is neuroprotective against beta-amyloid protein 25-35 (Abeta)-, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-, high density culture condition-, and serum deprivation-induced neuronal death in SK-N-SH human neuroblastoma cells. SK-N-SH cells express ERbeta, but not ERalpha, as detected by Western blot analysis. Among all the insults, MPTP, high density culture and serum deprivation induce apoptotic cell death in this cell system as detected by ELISA determination of mono/oligonucleosomes and DNA laddering, while Abeta induces necrotic cell death. The protective effects of E2 are abolished by the addition of tamoxifen and ICI 182,780 in the MPTP treated cells, but not in the other models, suggesting that the effect of E2 in the MPTP model is probably associated with activation of ERbeta. The addition of ICI 182,780 shows a mitogenic effect in SK-N-SH cells in the presence of E2 in control culture or in the Abeta treated groups. Also, ICI 182,780 induced expression of ERalpha. Collectively, the current studies suggest that E2 is neuroprotective in apoptotic and necrotic death induced by multiple insults in SK-N-SH human neuroblastoma cells. Involvement of ER is insult type dependent. ICI 182,780 is able to influence the expression of ERs, probably through upregulation of ERalpha when ERbeta is totally antagonized.
Collapse
Affiliation(s)
- Fang Ba
- Department of Physiology, Faculty of Medicine, University of Alberta, Edmonton, Alta T6G 2H7, Canada
| | | | | | | |
Collapse
|
68
|
Pongrac JL, Gibbs RB, Defranco DB. Estrogen-mediated regulation of cholinergic expression in basal forebrain neurons requires extracellular-signal-regulated kinase activity. Neuroscience 2004; 124:809-16. [PMID: 15026121 DOI: 10.1016/j.neuroscience.2004.01.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2004] [Indexed: 11/18/2022]
Abstract
Beyond the role estrogen plays in neuroendocrine feedback regulation involving hypothalamic neurons, other roles for estrogen in maintaining the function of CNS neurons remains poorly understood. Primary cultures of embryonic rat neurons together with radiometric assays were used to demonstrate how estrogen alters the cholinergic phenotype in basal forebrain by differentially regulating sodium-coupled high-affinity choline uptake and choline acetyltransferase activity. High-affinity choline uptake was significantly increased 37% in basal forebrain cholinergic neurons grown in the presence of a physiological dose of estrogen (5 nM) from 4 to 10 days in vitro whereas choline acetyltransferase activity was not significantly changed in the presence of 5 or 50 nM estrogen from 4 to 10 or 10 to 16 days in vitro. Newly-synthesized acetylcholine was significantly increased 35% following 6 days of estrogen treatment (10 days in vitro). These effects are in direct contrast to those found for nerve growth factor; that is, nerve growth factor can enhance the cholinergic phenotype through changes in choline acetyltransferase activity alone. This is most surprising given that mitogen-activated protein kinase and extracellular-signal-regulated kinase1/2, kinases also activated in the signaling pathway of nerve growth factor, were found to participate in the estrogen-mediated changes in the cholinergic phenotype. Likewise, general improvement in the viability of the cultures treated with estrogen does not account for the effects of estrogen as determined by lactate dehydrogenase release and nerve growth factor-responsiveness. These findings provide evidence that estrogen enhances the differentiated phenotype in basal forebrain cholinergic neurons through second messenger signaling in a manner distinct from nerve growth factor and independent of improved survival.
Collapse
Affiliation(s)
- J L Pongrac
- Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | | | |
Collapse
|
69
|
Carrer HF, Cambiasso MJ, Brito V, Gorosito S. Neurotrophic Factors and Estradiol Interact To Control Axogenic Growth in Hypothalamic Neurons. Ann N Y Acad Sci 2003; 1007:306-16. [PMID: 14993063 DOI: 10.1196/annals.1286.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Previous work from our laboratory has shown that in cultures of hypothalamic neurons obtained from male fetuses at embryonic day 16, the axogenic response to estrogen (E2) is contingent on coculture with target glia or target glia-conditioned media (CM). Neither the estrogen receptor blockers tamoxifen nor ICI 182,780 prevented the axogenic effects of the hormone. Estradiol made membrane-impermeable by conjugation to a protein of high molecular weight (E2-BSA) preserved its axogenic capacity, suggesting the possibility of a membrane effect responsible for the action of E2. Western blot analysis of extracts from homogenates of cultured neurons grown with E2 and CM from target glia had more TrkB than cultures with CM alone or E2 alone. To further investigate the interaction between E2 and the neurotrophin receptors, we used a specific antisense oligonucleotide (AS) to prevent the estradiol-induced increase of TrkB. The effect of E2 was suppressed in cultures in which TrkB was down-regulated by the AS, showing decreased axonal elongation when compared with neurons treated with E2 without AS or with sense TrkB. In cultures grown with AS, the axonal length of E2-treated cultures was not different from cultures without E2. Evidence suggesting cross-talk between E2 and neurotrophic factor(s) prompted investigation of signaling along the MAPK cascade. Immuno blotting of E2-treated cultures showed increased levels of phosphorylated ERK1 and ERK2. UO126 but not LY294002 blocked E2-induced axonal elongation, suggesting that the MAPKs are involved in this response.
Collapse
Affiliation(s)
- H F Carrer
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Casilla de Correo 389, 5000 Cordóba, Argentina.
| | | | | | | |
Collapse
|
70
|
Beyer C, Pawlak J, Brito V, Karolczak M, Ivanova T, Kuppers E. Regulation of Gene Expression in the Developing Midbrain by Estrogen: Implication of Classical and Nonclassical Steroid Signaling. Ann N Y Acad Sci 2003; 1007:17-28. [PMID: 14993036 DOI: 10.1196/annals.1286.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Estrogen plays an important role during midbrain development. This is indicated by the presence of nuclear estrogen receptors and the transient expression of the estrogen-forming enzyme aromatase. A number of recent studies have shown that estrogen promotes the differentiation and survival, as well as physiological performance, of midbrain dopaminergic cells. In addition, we have reported that both ways of cellular estrogen signaling (classical and nonclassical) as well as interactions with nonneuronal target cells are involved in the transmission of intra- and intercellular estrogen effects in this brain region. This study provides additional evidence that (i) estrogen is capable of regulating gene expression in cultured embryonic neurons and astrocytes differently and (ii) both signaling mechanisms, i.e., classically through nuclear receptors and nonclassically through the stimulation of membrane-estrogen receptors, which are coupled to distinct intracellular signal transduction cascades, contribute diversely to gene regulation. These data reveal a high degree of complexity of estrogen action at the genomic level in the developing brain. Further studies are warranted to unravel the exact contribution of the differently regulated genes for developmental estrogen action.
Collapse
Affiliation(s)
- Cordian Beyer
- Abteilung Anatomie und Zellbiologie, Universität Ulm, D-89069 Ulm, Germany.
| | | | | | | | | | | |
Collapse
|
71
|
Audesirk T, Cabell L, Kern M, Audesirk G. β-estradiol influences differentiation of hippocampal neurons in vitro through an estrogen receptor-mediated process. Neuroscience 2003; 121:927-34. [PMID: 14580943 DOI: 10.1016/s0306-4522(03)00294-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We utilized morphometric analysis of 3 day cultures of hippocampal neurons to determine the effects of both estradiol and the synthetic estrogen receptor modulator raloxifene on several parameters of neuronal growth and differentiation. These measurements included survival, neurite production, dendrite number, and axon and dendrite length and branching. 17 beta-Estradiol (10 nM) selectively stimulated dendrite branching; this effect was neither mimicked by alpha-estradiol, nor blocked by the estrogen receptor antagonist ICI 182780. The selective estrogen receptor modulator raloxifene (100 nM) neither mimicked nor reversed the effects of estradiol on dendritic branching. Western immunoblotting for the alpha and beta subtypes of estrogen receptor revealed the presence of alpha, but not beta, estrogen receptors in our hippocampal cultures. There is growing recognition of the effects of 17 beta-estradiol on neuronal development and physiology, with implications for brain sexual dimorphism, plasticity, cognition, and the maintenance of cognitive function during aging. The role of estradiol in hippocampal neuronal differentiation and function has particular implications for learning and memory. These data support the hypothesis that 17 beta-estradiol is acting via alpha estrogen receptors in influencing hippocampal development in vitro. Raloxifene, prescribed to combat osteoporosis in post-menopausal women, is a selective estrogen receptor modulator with tissue-specific agonist/antagonist properties. Because raloxifene had no effect on dendritic branching, we hypothesize that it does not interact with the alpha estrogen receptor in this experimental paradigm.
Collapse
Affiliation(s)
- T Audesirk
- Biology Department, University of Colorado at Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | | | | | | |
Collapse
|
72
|
Abstract
Oestrogen is important for the development of neuroendocrine centres and other neural networks including limbic and motor systems. Later in adulthood, oestrogen regulates the functional performance of different neural systems and is presumably implicated in the modulation of cognitive efficiency. Although still a matter of controversial discussion, clinical and experimental studies point at a potential neuroprotective role of oestrogen. Concerning the concept of cellular oestrogen action, it is undisputed that it comprises the binding and activation of nuclear receptors. The last decades have, however, immensely broadened the spectrum of steroid signalling within a cell. Novel steroid-activated intracellular signalling mechanisms were described which are usually termed 'non-classical' or 'non-genomic'. The brain appears to be a rich source of this new mode of oestrogen action. Studies from the past years have pinpointed non-classical oestrogen effects in many CNS regions. All available data support the view that non-classical oestrogen action requires interactions with putative membrane binding sites/receptors. In this article, we aim at compiling the most recent findings on the nature and identity of membrane oestrogen receptors with respect to the brain. We also attempt to turn readers attention to the coupling of these 'novel' receptors to distinct intracellular signalling pathways.
Collapse
Affiliation(s)
- Cordian Beyer
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Ulm, Germany.
| | | | | |
Collapse
|
73
|
Sato K, Matsuki N, Ohno Y, Nakazawa K. Estrogens inhibit l-glutamate uptake activity of astrocytes via membrane estrogen receptor alpha. J Neurochem 2003; 86:1498-505. [PMID: 12950458 DOI: 10.1046/j.1471-4159.2003.01953.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We investigated the effects of estrogen-related compounds including xenoestrogens [17beta-estradiol (E2), 17alpha-ethynylestradiol (EE), diethylstilbestrol (DES), p-nonylphenol (PNP), bisphenol A (BPA) and 17alpha-estradiol (17alpha)] on l-glu uptake by cultured astrocytes via glutamate-aspartate transporter (GLAST). After 24 h treatment, E2 inhibited the l-glu uptake at 1 micro m and higher concentrations. EE and DES also inhibited the l-glu uptake at 1 nm and higher concentrations. The other four compounds had no effect. The effects of E2, EE and DES were completely blocked by 10 nm of ICI182 780 (ICI). beta-Estradiol 17-hemisuccinate : bovine serum albumin (E2-BSA), a membrane-impermeable conjugate of E2, also elicited the inhibition of l-glu uptake at 1 nm and higher concentrations, and the effect was blocked by ICI. 16alpha-Iodo-17beta-estradiol (16alphaIE2), an estrogen receptor alpha (ERalpha) selective ligand, revealed an inhibitory effect at 10 nm, while genistein, an ERbeta selective ligand, failed to reveal such an effect at this concentration. Western blot analysis showed that the predominant ER of cultured astrocytes was ERalpha. The colocalization of ERalpha with GLAST on plasma membranes was immunohistochemically detected in these cells. From these results, we concluded that estrogens down-regulate l-glu uptake activity of astrocytes via membrane ERalpha.
Collapse
Affiliation(s)
- Kaoru Sato
- Division of Pharmacology, National Institute of Health Sciences, Kamiyoga, Setagaya-ku, Tokyo, Japan.
| | | | | | | |
Collapse
|
74
|
Losel RM, Falkenstein E, Feuring M, Schultz A, Tillmann HC, Rossol-Haseroth K, Wehling M. Nongenomic steroid action: controversies, questions, and answers. Physiol Rev 2003; 83:965-1016. [PMID: 12843413 DOI: 10.1152/physrev.00003.2003] [Citation(s) in RCA: 399] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Steroids may exert their action in living cells by several ways: 1). the well-known genomic pathway, involving hormone binding to cytosolic (classic) receptors and subsequent modulation of gene expression followed by protein synthesis. 2). Alternatively, pathways are operating that do not act on the genome, therefore indicating nongenomic action. Although it is comparatively easy to confirm the nongenomic nature of a particular phenomenon observed, e.g., by using inhibitors of transcription or translation, considerable controversy exists about the identity of receptors that mediate these responses. Many different approaches have been employed to answer this question, including pharmacology, knock-out animals, and numerous biochemical studies. Evidence is presented for and against both the participation of classic receptors, or proteins closely related to them, as well as for the involvement of yet poorly understood, novel membrane steroid receptors. In addition, clinical implications for a wide array of nongenomic steroid actions are outlined.
Collapse
Affiliation(s)
- Ralf M Losel
- Institut für klinische Pharmakologie, Klinikum Mannheim, Theodor-Kutzer-Ufer, D-68167 Mannheim, Germany
| | | | | | | | | | | | | |
Collapse
|
75
|
Kajta M, Beyer C. Cellular strategies of estrogen-mediated neuroprotection during brain development. Endocrine 2003; 21:3-9. [PMID: 12777697 DOI: 10.1385/endo:21:1:3] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2003] [Revised: 01/27/2003] [Accepted: 02/06/2003] [Indexed: 11/11/2022]
Abstract
The role of estrogen during brain development is well documented. Estrogen influences cell survival and differentiation and also controls the formation and maintenance of neural networks. Knowledge of trophic estrogen action in the central nervous system (CNS) was the basis for the establishment of research programs directed toward a potential function of estrogen as a neuroprotective factor in the adult brain. Considerable evidence has accumulated over the years supporting this hypothesis. Experimental and epidemiologic studies as well as clinical trials have demonstrated that estrogen is beneficial for the course of neurodegenerative disorders such as Parkinson and Alzheimer diseases but may also protect neurons from postischemic neuronal degeneration. In this article, we aim to unravel potential physiologic responses and cell survival strategies that allow a more detailed understanding of estrogen-mediated neuroprotection in the brain. In particular, we focus on the participation of estrogen in the regulation of apoptotic processes. Furthermore, we present data on reciprocal estrogen-growth factor interactions. Both of these mechanisms were found to operate during brain development and to conciliate estrogen effects on neurons. This makes them likely candidates for taking part in conveying estrogen-dependent neuroprotection in the adult CNS.
Collapse
Affiliation(s)
- Malgorzata Kajta
- Department of Endocrinology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | | |
Collapse
|
76
|
Mendez P, Azcoitia I, Garcia-Segura LM. Estrogen receptor alpha forms estrogen-dependent multimolecular complexes with insulin-like growth factor receptor and phosphatidylinositol 3-kinase in the adult rat brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 112:170-6. [PMID: 12670715 DOI: 10.1016/s0169-328x(03)00088-3] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Estradiol and insulin-like growth factor-I (IGF-I) have numerous functional interactions in the brain, including the regulation of neuroendocrine events, the control of reproductive behavior and the promotion of synaptic plasticity and neuronal survival. To explore the mechanisms involved in these interdependent actions of estradiol and IGF-I in the adult brain, the potential interactions of estrogen receptors with components of the IGF-I signaling system were assessed in this study. Systemic estradiol administration resulted in a transient immunocoprecipitation of the IGF-I receptor with the estrogen receptor alpha and in a transient increase in tyrosine phosphorylation of the IGF-I receptor in the hypothalamus of adult ovariectomized Wistar rats. Both effects were coincident in time, with a peak between 1 and 3 h after systemic estradiol administration. Three hours after estradiol treatment, there was an enhanced immunocoprecipitation of estrogen receptor alpha with p85 subunit of phosphatidylinositol 3-kinase, as well as an enhanced immunocoprecipitation of p85 with insulin receptor substrate-1. The interaction with the IGF-I receptor was specific for the alpha form of the estrogen receptor and was also induced by intracerebroventricular injection of IGF-I. These hormonal actions may be part of the mechanism by which estradiol activates IGF-I receptor signaling pathways in the brain and may explain the interdependence of estrogen receptors and the IGF-I receptor in synaptic plasticity, neuroprotection and other neural events.
Collapse
Affiliation(s)
- Pablo Mendez
- Instituto Cajal, C.S.I.C., Av. Dr. Arce 37, 28002, Madrid, Spain
| | | | | |
Collapse
|
77
|
Ravizza T, Galanopoulou AS, Velísková J, Moshé SL. Sex differences in androgen and estrogen receptor expression in rat substantia nigra during development: an immunohistochemical study. Neuroscience 2003; 115:685-96. [PMID: 12435407 DOI: 10.1016/s0306-4522(02)00491-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Gonadal hormones are important regulators of sexual differentiation of the CNS. Exposure to testosterone and estrogen during development causes permanent organizational differences between males and females. We previously described functional sex-related differences of the GABA(A)ergic circuits of the rat substantia nigra pars reticulata (SNR) involved in the control of flurothyl seizures. This sexual differentiation of the SNR is regulated by postnatal testosterone. To assess whether the organizing effects of testosterone in the SNR are mediated via the androgen receptor (AR) and/or estrogen receptors (ER), we used immunohistochemistry to study the ontogeny of AR, ERalpha and ERbeta expression in SNR and substantia nigra pars compacta (SNC) of male and female rats. Rats on the day of birth [postnatal day (PN) 0] and at PN1, PN5, PN15 and PN30 were used. AR- and ERbeta-immunopositive cells were present in SNR and SNC in both sexes and at all ages. ERalpha was not detected in male and female SNC at PN0-PN1. In both substantia nigra (SN) regions, there were developmentally regulated sex differences in AR, ERalpha and ERbeta immunoreactivity. In the SN, each receptor showed specific intracellular localization: AR was present in the nucleus, ERalpha and ERbeta were present both in nuclear and extranuclear compartments. ERalpha was detected also in processes. At PN0-PN1, quantitative analysis revealed sex and regional differences in the distribution of SN cells expressing AR and ERalpha, while ERbeta were equally present in both sexes. The presence of gonadal steroid receptors in the SN suggests that the biological effects of gonadal hormones in the CNS extend beyond reproduction-related functions and may affect and modify motor behaviors (including seizures) in a sex-specific manner. Based on the ontogeny of SNR ERbeta, we hypothesize that postnatal injections of testosterone may regulate the nigral GABA(A) system through the aromatization pathway and activation of ERbeta.
Collapse
Affiliation(s)
- T Ravizza
- Department of Neurology, Albert Einstein College of Medicine, K311, 1410 Pelham Parkway South, Bronx, NY, USA.
| | | | | | | |
Collapse
|
78
|
Golovine K, Schwerin M, Vanselow J. Three different promoters control expression of the aromatase cytochrome p450 gene (cyp19) in mouse gonads and brain. Biol Reprod 2003; 68:978-84. [PMID: 12604651 DOI: 10.1095/biolreprod.102.008037] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Aromatase cytochrome P450, the key enzyme of estrogen biosynthesis, is encoded by Cyp19. To elucidate the complex regulation of this gene in mouse gonads (ovary and testis) and brain (thalamic/hypothalamic areas), Cyp19 transcripts were isolated using rapid amplification of cDNA 5' ends and transcript concentrations were estimated in juveniles at different postnatal days (P0, P7, and P14) and in adult animals by real time polymerase chain reaction (PCR). In addition, the murine Cyp19 locus including all known exons and promoters was reconstructed from a recently published sequence of a mouse bacterial artificial chromosome. From each of the tissues investigated, Cyp19 transcripts with a specific 5' untranslated region (5' UTR) were isolated: T(ov) from ovary, T(br) from brain, and T(tes) from testis. T(tes) included a novel 5' UTR that did not show sequence similarities to other Cyp19 transcripts. Real time PCR experiments revealed similar levels of Cyp19 transcript concentrations in neonatal gonads of both sexes. The majority of transcripts were T(ov) in ovaries and T(tes) in testes. During further postnatal development, testicular Cyp19 transcript concentrations transiently decreased, but the contributions of different transcript variants basically remained unchanged. However, ovarian Cyp19 transcript concentrations increased by about 100 times, and almost 100% of all Cyp19 transcripts were identified as T(ov) in adult ovaries. Brains of both sexes showed highest transcript concentrations at P0. However, concentrations in female brains were reduced to adult levels earlier than in male brains. In brains of both sexes, T(br) was found to predominate throughout postnatal life. The results suggest that the mouse Cyp19 gene includes three different promoters that specifically direct expression in ovary, testis, and brain.
Collapse
Affiliation(s)
- Konstantin Golovine
- Research Unit Molecular Biology, Research Institute for the Biology of Farm Animals, 18196 Dummerstorf, Germany
| | | | | |
Collapse
|
79
|
Ivanova T, Beyer C. Estrogen regulates tyrosine hydroxylase expression in the neonate mouse midbrain. JOURNAL OF NEUROBIOLOGY 2003; 54:638-47. [PMID: 12555275 DOI: 10.1002/neu.10193] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Estrogen plays an important role during differentiation of midbrain dopaminergic neurons. This is indicated by the presence of estrogen receptors and the transient expression of the estrogen-forming enzyme aromatase within the dopaminergic cell groups. We have previously shown that estrogen regulates the plasticity of dopamine cells through the stimulation of neurite growth/arborization. In this study, we have analyzed the capability of estrogen to influence the activity of developing mouse dopamine neurons. The expression of tyrosine hydroxylase (TH) was assessed by competitive RT-PCR and Western blotting. The developmental expression of TH in the ventral midbrain was studied from embryonic day 15 until postnatal day 15 and revealed highest TH levels early postnatally. This profile coincides with the transient aromatase expression in this brain area. Using cultured midbrain cells, we found that estrogen increased TH mRNA/protein levels. The application of the estrogen receptor antagonist ICI 182,780 resulted in a complete inhibition of estrogen effects. To verify these data in vivo, fetuses were exposed in utero from E15 until birth to the aromatase inhibitor CGS 16949A or to CGS supplemented with estrogen. CGS caused a robust reduction in TH mRNA/protein levels in the midbrain, which could be restored by estrogen substitution. Taken together, our data strongly suggest that estrogen controls dopamine synthesis in the developing nigrostriatal dopaminergic system and support the concept that estrogen is implicated in the regulation of ontogenetic steps but also in the function of midbrain dopamine neurons.
Collapse
Affiliation(s)
- Tatiana Ivanova
- Abteilung Anatomie und Zellbiologie, Universität Ulm, D-89069 Ulm, Germany
| | | |
Collapse
|
80
|
Shibuya K, Takata N, Hojo Y, Furukawa A, Yasumatsu N, Kimoto T, Enami T, Suzuki K, Tanabe N, Ishii H, Mukai H, Takahashi T, Hattori TA, Kawato S. Hippocampal cytochrome P450s synthesize brain neurosteroids which are paracrine neuromodulators of synaptic signal transduction. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1619:301-16. [PMID: 12573490 DOI: 10.1016/s0304-4165(02)00489-0] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hippocampal pyramidal neurons and granule neurons of adult male rats are equipped with a complete machinery for the synthesis of pregnenolone, dehydroepiandrosterone, 17beta-estradiol and testosterone as well as their sulfate esters. These brain neurosteroids are synthesized by cytochrome P450s (P450scc, P45017alpha and P450arom) from endogenous cholesterol. Synthesis is acutely dependent on the Ca(2+) influx attendant upon neuron-neuron communication via N-methyl-D-aspartate (NMDA) receptors. Pregnenolone sulfate, estradiol and corticosterone rapidly modulate neuronal signal transduction and the induction of long-term potentiation via NMDA receptors and putative membrane steroid receptors. Brain neurosteroids are therefore promising neuromodulators that may either activate or inactivate neuron-neuron communication, thereby mediating learning and memory in the hippocampus.
Collapse
Affiliation(s)
- Keisuke Shibuya
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo at Komaba, Meguro, 153, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Cardona-Gómez GP, Mendez P, DonCarlos LL, Azcoitia I, Garcia-Segura LM. Interactions of estrogen and insulin-like growth factor-I in the brain: molecular mechanisms and functional implications. J Steroid Biochem Mol Biol 2002; 83:211-7. [PMID: 12650718 DOI: 10.1016/s0960-0760(02)00261-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In the brain, as in other tissues, estradiol interacts with growth factors. One of the growth factors that is involved in the neural actions of estradiol is insulin-like growth factor-I (IGF-I). Estradiol and IGF-I cooperate in the central nervous system to regulate neuronal development, neural plasticity, neuroendocrine events and the response of neural tissue to injury. The precise molecular mechanisms involved in these interactions are still not well understood. In the central nervous system there is abundant co-expression of estrogen receptors (ERs) and IGF-I receptors (IGF-IRs) in the same cells. Furthermore, the expression of estrogen receptors and IGF-I receptors in the brain is cross-regulated. In addition, using specific antibodies for the phosphorylated forms of extracellular-signal regulated kinase (ERK) 1 and ERK2 and Akt/protein kinase B (Akt/PKB) it has been shown that estradiol affects IGF-I signaling pathways in the brain. Estradiol treatment results in a dose-dependent increase in the phosphorylation of ERK and Akt/PKB in the brain of adult ovariectomized rats. In addition, estradiol and IGF-I have a synergistic effects on the activation of Akt/PKB in the adult rat brain. These findings suggest that estrogen effects in the brain may be mediated in part by the activation of the signaling pathways of the IGF-I receptor.
Collapse
|
82
|
Carrer HF, Cambiasso MJ. Sexual differentiation of the brain: genes, estrogen, and neurotrophic factors. Cell Mol Neurobiol 2002; 22:479-500. [PMID: 12585676 PMCID: PMC11533755 DOI: 10.1023/a:1021825317546] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Based on evidence obtained during the past 50 years, the current hypothesis to explain the sexual dimorphism of structure and function in the brain of vertebrates maintains that these differences are produced by the epigenetic action of gonadal hormones. However, evidence has progressively accumulated suggesting that genetic mechanisms controlling sexual-specific neuronal characteristics precede, or occur in parallel with, hormonal effects. 1. In cultures of hypothalamic neurons taken from gestation day 16 (GD16) embryos, treatment of sexually segregated cultures with estradiol (E2) induces axon growth in neurons from male neurons, but not from female neurons. In these cultures treatment with E2 increased the levels of tyrosine kinase type B (TrkB) and insulin-like growth factor I (IGF-I) receptors in male but not in female neurons. This and other sex differences cannot be explained by differences in hormonal environment, because the donor embryos were obtained when gonadal secretion of steroids is just beginning, before the perinatal surge of testosterone that determines development of the male brain beginning at GD17/18. 2. The response to estrogen is contingent upon coculture with heterotopic glia (mostly astrocytes) from a target region (amygdala) harvested from same-sex fetuses at GD16, whereas in the presence of homotopic glia or in cultures without glia, E2 had no effect. It was concluded that the axogenic effect of E2 depends on interaction between neurons and glia from a target region and that neurons from fetal male donors appear to mature earlier than neurons from females, a differentiated response that takes place prior to divergent exposure to gonadal secretions. 3. The effects of target and nontarget glia-conditioned media (CM) on the E2-induced growth of neuronal processes of hypothalamic neurons obtained from sexually segregated fetal donors were also studied. Estrogen added to media conditioned by target glia modified the number of primary neurites and the growth of axons of hypothalamic neurons of males but not of females. 4. Neither the Type III steroidal receptor blocker tamoxifen nor Type I antiestrogen ICI 182,780 prevented the axogenic effects of the hormone. Estradiol made membrane-impermeable by conjugation to a protein of high molecular weight (E2-BSA) preserved its axogenic capacity, suggesting the possibility of a membrane effect responsible for the action of E2. 5. Western blot analysis of the tyrosine kinase type A (TrkA), type B (TrkB), type C (TrkC), and insulin-like growth factor (IGF-I R) receptors in extracts from homogenates of cultured hypothalamic neurons showed that in cultures of male-derived neurons grown with E2 and CM from target glia, the amounts of TrkB and IGF-I R increased notably. Densitometric quantification showed that these cultures had more TrkB than cultures with CM alone or E2 alone. On the contrary, in cultures of female-derived neurons, the presence of CM alone induced maximal levels of TrkB, which were not further increased by E2; female-derived neurons in all conditions did not contain IGF-I R. Levels of TrkC were not modified by any experimental condition in male- or female-derived cultures and Trk A was not found in the homogenates. These results are compared with similar data from other laboratories and integrated in a model for the confluent interaction of estrogen and neurotrophic factors released by glia that may contribute to the sexual differentiation of the brain.
Collapse
Affiliation(s)
- Hugo F Carrer
- Instituto de Investigación Médica M. y M. Ferreyra, INIMEC-CONICET, Casilla de Correo 389, Córdoba 5000, Argentina.
| | | |
Collapse
|
83
|
Kumar A, Foster TC. 17beta-estradiol benzoate decreases the AHP amplitude in CA1 pyramidal neurons. J Neurophysiol 2002; 88:621-6. [PMID: 12163515 DOI: 10.1152/jn.2002.88.2.621] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Disruption of Ca(2+) homeostasis is hypothesized to mediate several electrophysiological markers of brain aging. Recent evidence indicates that estradiol can rapidly alter Ca(2+)-dependent processes in neurons through nongenomic mechanisms. In the current study, electrophysiological effects of 17beta-estradiol benzoate (EB) on the Ca(2+)-activated afterhyperpolarization (AHP) were investigated using intracellular sharp electrode recording in hippocampal slices from ovariectomized Fischer 344 female rats. The AHP amplitude was enhanced in aged (22-24 mo) compared with young (5-8 mo) rats and direct application of EB (100 pM) reduced the AHP in aged rats. The age-related difference was due, in part, to the increased AHP amplitude of aged animals, since an EB-mediated decrease in the AHP could be observed in young rats when the extracellular Ca(2+) was elevated to increase the AHP amplitude. In aged rats, bath application of EB occluded the ability of the L-channel blocker, nifedipine (10 microM), to attenuate the AHP. The results support a role for EB in modifying hippocampal Ca(2+)-dependent processes in a manner diametrically opposite that observed during aging, possibly through L-channel inhibition.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, College of Medicine, Lexington 40536, USA
| | | |
Collapse
|
84
|
Beyer C, Ivanova T, Karolczak M, Küppers E. Cell type-specificity of nonclassical estrogen signaling in the developing midbrain. J Steroid Biochem Mol Biol 2002; 81:319-25. [PMID: 12361721 DOI: 10.1016/s0960-0760(02)00119-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Estrogens have widespread biological functions in the CNS involving the coordination of developmental processes, the regulation of cell physiology, and the control of neuroendocrine systems. In the midbrain, estrogens promote the survival, maturation, and function of neurons and, in particular, of dopamine cells. Aside from classical signaling through nuclear estrogen receptors, we have provided evidence that cellular transmission of estrogen effects in the midbrain comprises a complex intracellular signaling scenario. The major conclusion drawn from our studies is that estrogens interact with yet unidentified membrane receptor complexes which stimulate the phospholipase C and induce the formation of inosite-tri-phosphate (IP(3)). This causes a rapid and transitory rise in intracellular free calcium. The modulation of calcium homeostasis is the primary nonclassical physiological response to estrogens in all cell types. Surprisingly, a different secondary downstream signaling cascade seems to be activated in each estrogen-responsive cell population, i.e. phosphatidylinositol-3 kinase (PI3-kinase) in GABAergic and cAMP/ protein kinase A (PKA) in dopaminergic neurons, mitogen-activated protein kinase (MAP-kinase) in astrocytes. The precise biological role of estrogens for the different cell types is still fragmentary. We assume that estrogens positively influence intracellular signaling mechanisms which are important for cell differentiation and survival. It remains to be elucidated what determines the cell type-specificity of these estrogen responses.
Collapse
Affiliation(s)
- Cordian Beyer
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Germany.
| | | | | | | |
Collapse
|
85
|
Abstract
Increasing evidence suggests that estrogens may protect the nigrostriatal dopaminergic pathway affected in Parkinson's disease (PD). Animal studies show that estrogens influence the synthesis, release, and metabolism of dopamine and can modulate dopamine receptor expression and function. Some clinical studies suggest that PD symptoms may be exacerbated after menopause and delayed or alleviated with hormone replacement therapy, but others have failed to observe positive estrogenic effects. The conflicting findings suggest that several variables, including age, estrogen dose and formulation, and timing and length of dosing period, may determine whether benefits are seen and the nature of these benefits. Further investigation is therefore needed for the relationship between estrogens and the nigrostriatal dopaminergic system.
Collapse
Affiliation(s)
- Lisa M Shulman
- The Rosalyn Newman Scholar of Clinical Research in Parkinson's Disease, University of Maryland School of Medicine, 22 South Greene Street N4W46, Baltimore, MD 21201, USA.
| |
Collapse
|
86
|
Sutter-Dub MT. Rapid non-genomic and genomic responses to progestogens, estrogens, and glucocorticoids in the endocrine pancreatic B cell, the adipocyte and other cell types. Steroids 2002; 67:77-93. [PMID: 11755172 DOI: 10.1016/s0039-128x(01)00142-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rapid biologic responses to injected steroids were described as early as 60 years ago. More recently, evidence has been presented that 17beta-estradiol given i.v. will double the uterine cAMP activity within 15 s (Proc Natl Acad Sci USA 1967;58:1711-8), and also that estrogens will bind to the outer surfaces of endometrial cells (Nature 1977;265:69-72), suggesting that these steroids can both engage and direct intracellular events. Unfortunately, studies of such rapid membrane effects of steroids have languished due to the accumulation of compelling data for the more slowly manifest actions of these compounds at the level of nuclear DNA. We report a number of observations in women, in experimental animals, and in isolated organ or cell systems using 17beta-estradiol, progesterone or glucocorticoids which provide ample evidence for rapid intracellular metabolic responses to these steroids, mediated by their actions at the cellular plasma membrane. Such rapid responses have been shown in various classic targets or not, such as the B cell of the endocrine pancreas and the fat cell. They involve plasma membrane binding, changes in membrane electrical activity, Ca2+ handling, G and Ras proteins, cAMP, cGMP, IP(3), DAG, phosphodiesterases, protein kinases, tyrosine kinases, ER kinases, and mitogen activated protein kinases (MAPks) and nitric oxide synthase. These recent findings are discussed in detail and should lead to a fuller understanding of the cellular effects of the steroid hormones.
Collapse
Affiliation(s)
- Marie Thérèse Sutter-Dub
- Université Bordeaux I, UFR de Biologie, Laboratoire d'Endocrinologie cellulaire: Mécanismes d'action d'hormones stéroides, Avenue des Facultés, F-33405 Talence Cedex, France.
| |
Collapse
|
87
|
Ivanova T, Mendez P, Garcia-Segura LM, Beyer C. Rapid stimulation of the PI3-kinase/Akt signalling pathway in developing midbrain neurones by oestrogen. J Neuroendocrinol 2002; 14:73-9. [PMID: 11903815 DOI: 10.1046/j.0007-1331.2001.00742.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Oestrogen promotes the differentiation of neurones in the central nervous system. In the rodent midbrain, the maturation of dopaminergic neurones appears to be under oestrogen control. This is supported by the fact that dopaminergic cells contain nuclear oestrogen receptors-alpha/beta (ER). Second, aromatase is transiently expressed in the developing midbrain. In previous studies, we have shown that oestrogen increases dopamine synthesis and plasticity of dopamine cells. These effects are transmitted through classical nuclear ER but require also the stimulation of nonclassical signalling pathways involving the activation of membrane receptors. This study attempted to identify nonclassical oestrogen-dependent signalling cascades which might be stimulated downstream of membrane ERs. Using cultured mouse midbrain cells, we could demonstrate by Western blotting, that oestrogen rapidly phosphorylates Akt, a kinase which is implicated in the phosphatidylinositol 3 (PI3)-kinase pathway. This effect was only seen in midbrain neurones but not astrocytes. Oestrogen-induced Akt phosphorylation was time- and dose-dependent, showing highest responses after 30 min and at a steroid concentration of 10(-8) and 10(-6) M. Immunocytochemistry for phosphorylated Akt (pAkt) demonstrated that pAkt is predominantly found in a nuclear/perinuclear position and that oestrogen exposure increased the number of pAkt-positive cells. To investigate the mechanisms which are involved in transmitting oestrogen effects on the cellular level, cells were treated with antagonists for distinct signalling pathways. The application of the nuclear ER antagonist ICI 182 780 did not abolish the oestrogen-induced Akt phosphorylation. In contrast, interrupting intracellular calcium signalling with BAPTA completely prevented this effect. The PI3-kinase inhibitor LY294002 also inhibited the activation of Akt by oestrogen. Our study clearly indicates that oestrogen can rapidly stimulate the PI3-kinase/Akt signalling cascade in differentiating midbrain neurones. This effect requires the intermediate activation of calcium-dependent signalling pathways. In conclusion, oestrogen effects in the developing midbrain appear to be connected with the PI3-kinase/Akt signalling mechanism.
Collapse
Affiliation(s)
- T Ivanova
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Ulm, Germany
| | | | | | | |
Collapse
|
88
|
Cambiasso MJ, Carrer HF. Nongenomic mechanism mediates estradiol stimulation of axon growth in male rat hypothalamic neurons in vitro. J Neurosci Res 2001; 66:475-81. [PMID: 11746365 DOI: 10.1002/jnr.1238] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The purpose of the present work was to investigate the participation of estradiol receptors (ER) in estrogen-induced axon growth in vitro. Hypothalamic neurons from 16 day (E16) male rat fetuses were cultured with or without 17-beta-estradiol at 1 x 10(-7) M in basal medium or medium conditioned by astroglia derived from ventral mesencephalon (CM). After 48 hr in vitro, neurite outgrowth was quantified by morphometric analysis. An axogenic effect could be demonstrated for estradiol added to CM. With RT-PCR, the mRNA transcript for ERalpha was found in the donor tissues as well as in the neuron cultures. In this model two specific nuclear ER blockers (tamoxifen and ICI 182,780) were ineffective in blocking the neuritogenic effect, and a membrane-impermeable estrogen-albumin construct (E2-BSA) was as effective as estradiol. These results indicate that the axogenic effect of estradiol at E16 is not exerted through the classical intracellular receptor signal transduction system and suggest the possibility of a membrane-mediated mechanism. The data are discussed in light of our previous findings pointing to the interdependent activation of the estrogenic and the trophic factor signaling pathways that mediate stimulated axon growth.
Collapse
Affiliation(s)
- M J Cambiasso
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Córdoba, Argentina
| | | |
Collapse
|
89
|
Cardona-Gómez GP, Mendez P, DonCarlos LL, Azcoitia I, Garcia-Segura LM. Interactions of estrogens and insulin-like growth factor-I in the brain: implications for neuroprotection. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2001; 37:320-34. [PMID: 11744097 DOI: 10.1016/s0165-0173(01)00137-0] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Data from epidemiological studies suggest that the decline in estrogen following menopause could increase the risk of neurodegenerative diseases. Furthermore, experimental studies on different animal models have shown that estrogen is neuroprotective. The mechanisms involved in the neuroprotective effects of estrogen are still unclear. Anti-oxidant effects, activation of different membrane-associated intracellular signaling pathways, and activation of classical nuclear estrogen receptors (ERs) could contribute to neuroprotection. Interactions with neurotrophins and other growth factors may also be important for the neuroprotective effects of estradiol. In this review we focus on the interaction between insulin-like growth factor-I (IGF-I) and estrogen signaling in the brain and on the implications of this interaction for neuroprotection. During the development of the nervous system, IGF-I promotes the differentiation and survival of specific neuronal populations. In the adult brain, IGF-I is a neuromodulator, regulates synaptic plasticity, is involved in the response of neural tissue to injury and protects neurons against different neurodegenerative stimuli. As an endocrine signal, IGF-I represents a link between the growth and reproductive axes and the interaction between estradiol and IGF-I is of particular physiological relevance for the regulation of growth, sexual maturation and adult neuroendocrine function. There are several potential points of convergence between estradiol and IGF-I receptor (IGF-IR) signaling in the brain. Estrogen activates the mitogen-activated protein kinase (MAPK) pathway and has a synergistic effect with IGF-I on the activation of Akt, a kinase downstream of phosphoinositol-3 kinase. In addition, IGF-IR is necessary for the estradiol induced expression of the anti-apoptotic molecule Bcl-2 in hypothalamic neurons. The interaction of ERs and IGF-IR in the brain may depend on interactions between neural cells expressing ERs with neural cells expressing IGF-IR, or on direct interactions of the signaling pathways of alpha and beta ERs and IGF-IR in the same cell, since most neurons expressing IGF-IR also express at least one of the ER subtypes. In addition, studies on adult ovariectomized rats given intracerebroventricular (i.c.v.) infusions with antagonists for ERs or IGF-IR or with IGF-I have shown that there is a cross-regulation of the expression of ERs and IGF-IR in the brain. The interaction of estradiol and IGF-I and their receptors may be involved in different neural events. In the developing brain, ERs and IGF-IR are interdependent in the promotion of neuronal differentiation. In the adult, ERs and IGF-IR interact in the induction of synaptic plasticity. Furthermore, both in vitro and in vivo studies have shown that there is an interaction between ERs and IGF-IR in the promotion of neuronal survival and in the response of neural tissue to injury, suggesting that a parallel activation or co-activation of ERs and IGF-IR mediates neuroprotection.
Collapse
Affiliation(s)
- G P Cardona-Gómez
- Instituto Cajal, C.S.I.C., Avenida Doctor Arce 37, E-28002, Madrid, Spain
| | | | | | | | | |
Collapse
|
90
|
Ivanova T, Küppers E, Engele J, Beyer C. Estrogen stimulates brain-derived neurotrophic factor expression in embryonic mouse midbrain neurons through a membrane-mediated and calcium-dependent mechanism. J Neurosci Res 2001; 66:221-30. [PMID: 11592117 DOI: 10.1002/jnr.1214] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We have provided evidence that 17beta-estradiol (E) synthesized in the midbrain promotes the differentiation of midbrain dopamine neurons through nonclassical steroid action. Because these developmental effects resemble those reported for brain-derived neurotrophic factor (BDNF), we hypothesized that E influences dopaminergic cell differentiation through a BDNF-dependent mechanism. Competitive RT-PCR and ELISA techniques were employed to study first the developmental pattern of BDNF and trkB expression in the mouse midbrain. BDNF protein/mRNA levels peaked postnatally, whereas trkB did not fluctuate perinatally. To prove the hypothesis that E regulates BDNF expression in vivo, fetuses and newborns were treated with the aromatase antagonist CGS 16949A. CGS 16949A exposure reduced midbrain BDNF mRNA/protein levels. The coapplication of CGS 16949A and E abolished this effect. Midbrain cultures from mouse fetuses were used to investigate intracellular signaling mechanisms involved in transmitting E effects. Estrogen increased expression of BDNF but not of other neurotrophins. As concerns the related signaling mechanism, these effects were antagonized by interrupting intracellular Ca(2+) signaling with BAPTA and thapsigargin but not by the estrogen receptor antagonist ICI 182,780. Insofar as E effects on BDNF mRNA expression were inhibited by cycloheximide, it appears likely that other, not yet characterized intermediate proteins take part in the estrogenic regulation of BDNF expression. We conclude that E exerts its stimulatory effect on the differentiation of dopaminergic neurons by coordinating BDNF expression. This particular E effect appears to be transmitted through Ca(2+)-dependent signaling cascades upon activation of putative membrane estrogen receptors.
Collapse
Affiliation(s)
- T Ivanova
- Abteilung Anatomie und Zellbiologie, Universität Ulm, 89069 Ulm, Germany
| | | | | | | |
Collapse
|
91
|
Küppers E, Ivanova T, Karolczak M, Lazarov N, Föhr K, Beyer C. Classical and nonclassical estrogen action in the developing midbrain. Horm Behav 2001; 40:196-202. [PMID: 11534982 DOI: 10.1006/hbeh.2001.1671] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
There is widespread acceptance that estrogen is involved in various steps of cellular differentiation during brain development. In the past years, we have demonstrated such a developmental role for estrogen in the rodent midbrain. Precisely, estrogen affects midbrain dopamine neurons with respect to functional and morphological maturation. On the cellular level, estrogen may act classically by binding and activating its respective nuclear receptors, thereby controlling the transcription of target genes. On the other hand, many estrogen effects in the CNS are transmitted nonclassically by interactions with putative membrane receptors and by stimulating distinct intracellular signaling cascades. In the midbrain, classical and nonclassical estrogen signaling routes operate side by side to ensure the proper development of dopaminergic cells. In the present report, we detail some of the cellular and molecular events which are activated by estrogen and are thought to take part in the estrogen-mediated stimulation of dopamine neuron differentiation.
Collapse
Affiliation(s)
- E Küppers
- Anatomy and Cell Biology, University of Ulm, D-89069 Ulm, Germany
| | | | | | | | | | | |
Collapse
|
92
|
Lee SJ, McEwen BS. Neurotrophic and neuroprotective actions of estrogens and their therapeutic implications. Annu Rev Pharmacol Toxicol 2001; 41:569-91. [PMID: 11264469 DOI: 10.1146/annurev.pharmtox.41.1.569] [Citation(s) in RCA: 400] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Originally known for its regulation of reproductive functions, estradiol, a lipophilic hormone that can easily cross plasma membranes as well as the blood-brain barrier, maintains brain systems subserving arousal, attention, mood, and cognition. In addition, both synthetic and natural estrogens exert neurotrophic and neuroprotective effects. There is increasing evidence that estrogen actions are mediated by nongenomic as well as direct and indirect genomic pathways. Although in vitro models have provided the most extensive evidence for neurotrophic and neuroprotective actions to date, there are also in vivo studies that support these actions.
Collapse
Affiliation(s)
- S J Lee
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York 10021, USA.
| | | |
Collapse
|
93
|
Differential mechanisms of neuroprotection by 17 beta-estradiol in apoptotic versus necrotic neurodegeneration. J Neurosci 2001. [PMID: 11306613 DOI: 10.1523/jneurosci.21-08-02600.2001] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The major goal of this study was to compare mechanisms of the neuroprotective potential of 17 beta-estradiol in two models for oxidative stress-independent apoptotic neuronal cell death with that in necrotic neuronal cell death in primary neuronal cultures derived from rat hippocampus, septum, or cortex. Neuronal apoptosis was induced either by staurosporine or ethylcholine aziridinium (AF64A), as models for necrotic cell death glutamate exposure or oxygen-glucose deprivation (OGD) were applied. Long-term (20 hr) pretreatment (0.1 microm 17 beta-estradiol) was neuroprotective in apoptotic neuronal cell death induced by AF64A (40 microm) only in hippocampal and septal neuronal cultures and not in cortical cultures. The neuroprotective effect was blocked by the estrogen antagonists ICI 182,780 and tamoxifen and the phosphatidylinositol 3-kinase (PI3-K) inhibitor LY294002. In glutamate and OGD-induced neuronal damage, long-term pretreatment was not effective. In contrast, short-term (1 hr) pretreatment with 17 beta-estradiol in the dose range of 0.5-1.0 microm significantly reduced the release of lactate dehydrogenase and improved morphology of cortical cultures exposed to glutamate or OGD but was not effective in the AF64A model. Staurosporine-induced apoptosis was not prevented by either long- or short-term pretreatment. The strong expression of the estrogen receptor-alpha and the modulation of Bcl proteins by 17 beta-estradiol in hippocampal and septal but not in cortical cultures indicates that the prevention of apoptotic, but not of necrotic, neuronal cell death by 17 beta-estradiol possibly depends on the induction of Bcl proteins and the density of estrogen receptor-alpha.
Collapse
|
94
|
Kritzer MF, Pugach I. Administration of tamoxifen but not flutamide to hormonally intact, adult male rats mimics the effects of short-term gonadectomy on the catecholamine innervation of the cerebral cortex. J Comp Neurol 2001; 431:444-59. [PMID: 11223814 DOI: 10.1002/1096-9861(20010319)431:4<444::aid-cne1082>3.0.co;2-m] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Gonadectomy in adult male rats induces a series of changes in cortical catecholamine innervation that begins with a large, but transient decrease in the density of tyrosine hydroxylase- but not dopamine-beta-hydroxylase-immunoreactive axons in sensory, motor, and association cortices. More recent studies have shown that estradiol maintains these presumed dopamine afferents but that supplementing acutely gonadectomized rats with dihydrotestosterone provides no protective effects for innervation. These findings suggest that the depression of mesocortical dopamine axons that follows gonadectomy is stimulated by changes in estrogen signaling. The studies presented here examined tyrosine hydroxylase and dopamine-beta-hydroxylase innervation in hormonally intact adult male rats treated for 4 days with the nonsteroidal antiestrogen tamoxifen or with the nonsteroidal antiandrogen flutamide to probe for additional evidence for this selective hormone sensitivity and for insights into the intracellular mechanisms that may govern it. Qualitative and quantitative comparisons of innervation with corresponding data from control and acutely gonadectomized rats revealed that administration of the antiestrogen tamoxifen in hormonally intact rats produced deficits in catecholamine innervation that mirrored those induced by short-term gonadectomy. The antiandrogen flutamide, however, had no discernible impact on cortical afferents. When considered within the context of the known pharmacology and sites of action of tamoxifen, these findings not only provide additional support for an initial phase of selective estrogen sensitivity among the cortical catecholamines but also suggest that it is stimulation of intracellular estrogen receptors that confers this sensitivity in the adult rat cerebrum.
Collapse
Affiliation(s)
- M F Kritzer
- Department of Neurobiology and Behavior, SUNY at Stony Brook, Stony Brook, New York 11794-5230, USA.
| | | |
Collapse
|
95
|
Ivanova T, Karolczak M, Beyer C. Estrogen stimulates the mitogen-activated protein kinase pathway in midbrain astroglia. Brain Res 2001; 889:264-9. [PMID: 11166717 DOI: 10.1016/s0006-8993(00)03149-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Estrogen stimulates the development of midbrain dopamine neurons predominantly by acting through membrane receptors coupled to Ca(2+)-signaling. In this report, we describe that estrogen activates extracellular signal-regulated kinases (ERK1/2) in midbrain astrocytes but not neurons. This effect was inhibited by BAPTA which interrupts Ca(2+)-signaling but not by antagonists specific for other signaling pathways. The activation of the MAP kinase pathway suggests a potential role for astrocytes in mediating estrogen effects in the midbrain.
Collapse
Affiliation(s)
- T Ivanova
- Abteilung Anatomie und Zellbiologie, Universität Ulm, D-89069, Ulm, Germany
| | | | | |
Collapse
|
96
|
Abstract
This review highlights recent evidence from clinical and basic science studies supporting a role for estrogen in neuroprotection. Accumulated clinical evidence suggests that estrogen exposure decreases the risk and delays the onset and progression of Alzheimer's disease and schizophrenia, and may also enhance recovery from traumatic neurological injury such as stroke. Recent basic science studies show that not only does exogenous estradiol decrease the response to various forms of insult, but the brain itself upregulates both estrogen synthesis and estrogen receptor expression at sites of injury. Thus, our view of the role of estrogen in neural function must be broadened to include not only its function in neuroendocrine regulation and reproductive behaviors, but also to include a direct protective role in response to degenerative disease or injury. Estrogen may play this protective role through several routes. Key among these are estrogen dependent alterations in cell survival, axonal sprouting, regenerative responses, enhanced synaptic transmission and enhanced neurogenesis. Some of the mechanisms underlying these effects are independent of the classically defined nuclear estrogen receptors and involve unidentified membrane receptors, direct modulation of neurotransmitter receptor function, or the known anti-oxidant activities of estrogen. Other neuroprotective effects of estrogen do depend on the classical nuclear estrogen receptor, through which estrogen alters expression of estrogen responsive genes that play a role in apoptosis, axonal regeneration, or general trophic support. Yet another possibility is that estrogen receptors in the membrane or cytoplasm alter phosphorylation cascades through direct interactions with protein kinases or that estrogen receptor signaling may converge with signaling by other trophic molecules to confer resistance to injury. Although there is clear evidence that estradiol exposure can be deleterious to some neuronal populations, the potential clinical benefits of estrogen treatment for enhancing cognitive function may outweigh the associated central and peripheral risks. Exciting and important avenues for future investigation into the protective effects of estrogen include the optimal ligand and doses that can be used clinically to confer benefit without undue risk, modulation of neurotrophin and neurotrophin receptor expression, interaction of estrogen with regulated cofactors and coactivators that couple estrogen receptors to basal transcriptional machinery, interactions of estrogen with other survival and regeneration promoting factors, potential estrogenic effects on neuronal replenishment, and modulation of phenotypic choices by neural stem cells.
Collapse
|
97
|
Kritzer MF. Effects of acute and chronic gonadectomy on the catecholamine innervation of the cerebral cortex in adult male rats: Insensitivity of axons immunoreactive for dopamine‐β‐hydroxylase to gonadal steroids, and differential sensitivity of axons immunoreactive for tyrosine hydroxylase to ovarian and testicular hormones. J Comp Neurol 2000. [DOI: 10.1002/1096-9861(20001127)427:4<617::aid-cne9>3.0.co;2-#] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Mary F. Kritzer
- Department of Neurobiology and Behavior, SUNY at Stony Brook, Stony Brook, New York 11794‐5230
| |
Collapse
|
98
|
Abstract
In addition to their role as sex hormones, it has been known for many years that oestrogens have protective effects on the vasculature. These have been implicated in the reduced incidence of cardiovascular disorders in premenopausal women and in post-menopausal women receiving oestrogen replacement therapy. This protection has been found to be due, in part at least, to direct effects of oestrogens on blood vessels. This review will summarize the available literature regarding oestrogenic effects on vascular contractility. Two major influences of oestrogens will be discussed; first the genomic effects induced by chronic administration of steroid hormones, and second, the rapid effects on vascular smooth muscle by non-genomic, and as yet not fully identified, mechanisms. In so doing, the diversity of oestrogenic actions on vascular contractility will be highlighted and the protective role of these agents against adverse cardiovascular events discussed.
Collapse
Affiliation(s)
- C E Austin
- Department of Medicine, Manchester Royal Infirmary, UK.
| |
Collapse
|
99
|
Israel JM, Poulain DA. 17-Oestradiol modulates in vitro electrical properties and responses to kainate of oxytocin neurones in lactating rats. J Physiol 2000; 524 Pt 2:457-70. [PMID: 10766926 PMCID: PMC2269881 DOI: 10.1111/j.1469-7793.2000.t01-2-00457.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
1. Intracellular current clamp recordings were performed from identified oxytocin (OT) neurones in acute hypothalamic slices taken from lactating Wistar rats at early (5th day: LD-5) and late (21st day: LD-21) lactation. 2. The basic electrophysiological properties of LD-21 OT neurones differed from those of LD-5 OT neurones: their resting membrane potential was more depolarised (-51.5 versus -54.9 mV); their action potential duration was longer (1.6 versus 1.2 ms); their hyperpolarising after-potential (HAP) following single spikes and after-hyperpolarisation (AHP) following a burst of action potentials had smaller amplitudes (-46 and -67 %, respectively); and they lacked spike frequency adaptation during a burst. 3. In LD-21 neurones bath application of 17beta-oestradiol (10-7 M, 6-14 min) reversibly restored all these properties to values observed in LD-5 cells. This treatment had no effect on LD-5 neurones. 4. LD-21 neurones were less sensitive to kainate than LD-5 neurones. 17beta-Oestradiol significantly potentiated the kainate-induced response in LD-21, but not in LD-5 neurones. 5. The effects of 17beta-oestradiol were presumably mediated through a non-genomic mechanism since they occurred within a few minutes of administration, and disappeared within 30-40 min of washout. They were not inhibited by tamoxifen, an antagonist of the nuclear oestrogen receptor ER-alpha. Lastly, cholesterol, a non-active lipophilic molecule, had no effect. 6. Our observations demonstrate that, in the absence of 17beta-oestradiol, the basic electrical properties and sensitivity to kainate of OT neurones become altered between early and late lactation. However, the rise in circulating levels of oestrogens during the late phase of lactation may contribute to maintain OT neurone reactivity as long as suckling continues.
Collapse
Affiliation(s)
- J M Israel
- Laboratoire de Neurobiologie Morphofonctionnelle, INSERM U 378, Universite Victor Segalen Bordeaux 2, 1 rue Camille Saint-Saëns, 33077 Bordeaux cedex, France.
| | | |
Collapse
|
100
|
Shaw L, Taggart MJ, Austin C. Mechanisms of 17 beta-oestradiol induced vasodilatation in isolated pressurized rat small arteries. Br J Pharmacol 2000; 129:555-65. [PMID: 10711355 PMCID: PMC1571867 DOI: 10.1038/sj.bjp.0703084] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. The influence of 17 beta-oestradiol on pressurized isolated rat mesenteric and coronary small arteries was investigated. 2. 17 beta-oestradiol caused rapid (t1.0 < 5 mins) concentration-dependent relaxations of pre-contracted pressurized (50 mmHg) isolated rat mesenteric and coronary arteries. Similar responses were observed in both vessel types. Significant relaxations were only observed at concentrations exceeding 3 microM. 3. The vasodilatory responses in both types of artery were unaffected by 10 microM L-nitro arginine (L-NNA) alone or in the presence of 10 microM indomethacin, inhibitors of nitric oxide and prostaglandin synthesis respectively. They were also unaffected by the pre-contracting agent used i.e. high K+ or U46619 (a thromboxane analogue). 4. Neither the oestrogen receptor antagonist ICI 182,780 (10 microM) nor the protein synthesis inhibitor cycloheximide (100 microM) had any effect on the responses of mesenteric arteries to 17 beta-oestradiol. 5. 17 alpha-oestradiol had only a minor effect on mesenteric arterial diameter over a concentration range similar to the effective vasodilatory range for 17 beta-oestradiol. 6. Membrane impermeant 17 beta-oestradiol conjugated to bovine serum albumin (beta-oestradiol-17-hemisuccinate-BSA) (E-H-BSA) resulted in a vasodilatation of pressurized arteries. 7. Wortmannin, an inhibitor of myosin light chain kinase, near maximally relaxed pressurized mesenteric arteries although the time course for the response was significantly slower than that for 17 beta-oestradiol. 8. These results taken together suggest that the acute effects of 17 beta-oestradiol on isolated pressurized arterial tone may be due to effects directly on the vascular smooth muscle via non-genomic mechanisms that involve a stereospecific interaction at the plasma membrane.
Collapse
Affiliation(s)
- L Shaw
- University Department of Medicine, Manchester Royal Infirmary
| | | | | |
Collapse
|