51
|
Morale MC, L'Episcopo F, Tirolo C, Giaquinta G, Caniglia S, Testa N, Arcieri P, Serra PA, Lupo G, Alberghina M, Harada N, Honda S, Panzica GC, Marchetti B. Loss of aromatase cytochrome P450 function as a risk factor for Parkinson's disease? ACTA ACUST UNITED AC 2007; 57:431-43. [PMID: 18063054 DOI: 10.1016/j.brainresrev.2007.10.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Revised: 10/24/2007] [Accepted: 10/26/2007] [Indexed: 12/21/2022]
Abstract
The final step in the physiological synthesis of 17beta estradiol (E(2)) is aromatization of precursor testosterone by a CYP19 gene product, cytochrome P450 estrogen aromatase in the C19 steroid metabolic pathway. Within the central nervous system (CNS) the presence, distribution, and activity of aromatase have been well characterized. Developmental stage and injury are known modulators of brain enzyme activity, where both neurons and glial cells reportedly have the capability to synthesize this key estrogenic enzyme. The gonadal steroid E(2) is a critical survival, neurotrophic and neuroprotective factor for dopaminergic neurons of the substantia nigra pars compacta (SNpc), the cells that degenerate in Parkinson's disease (PD). In previous studies we underlined a crucial role for the estrogenic status at the time of injury in dictating vulnerability to the parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our ongoing studies address the contribution of brain aromatase and extragonadal E(2) as vulnerability factors for PD pathology in female brain, by exposing aromatase knockout (ArKO, -/-) female mice which are unable to synthesize estrogens to MPTP. Our initial results indicate that aromatase deficiency from early embryonic life significantly impairs the functional integrity of SNpc tyrosine hydroxylase-positive neurons and dopamine transporter innervation of the caudate-putamen in adulthood. In addition, ArKO females exhibited a far greater vulnerability to MPTP-induced nigrostriatal damage as compared to their Wt type gonadally intact and gonadectomized counterparts. Characterization of this novel implication of P450 aromatase as determining factor for PD vulnerability may unravel new avenues for the understanding and development of novel therapeutic approaches for Parkinson's disease.
Collapse
Affiliation(s)
- M C Morale
- OASI Institute for Research and Care on Mental Retardation and Brain Aging (IRCCS), Neuropharmacology Section, 94018 Troina, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Noell S, Fallier-Becker P, Beyer C, Kröger S, Mack AF, Wolburg H. Effects of agrin on the expression and distribution of the water channel protein aquaporin-4 and volume regulation in cultured astrocytes. Eur J Neurosci 2007; 26:2109-18. [PMID: 17927773 DOI: 10.1111/j.1460-9568.2007.05850.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Agrin is a heparan sulfate proteoglycan of the extracellular matrix and is known for organizing the postsynaptic differentiation of the neuromuscular junction. Increasing evidence also suggests roles for agrin in the developing CNS, including the formation and maintenance of the blood-brain barrier. Here we describe effects of agrin on the expression and distribution of the water channel protein aquaporin-4 (AQP4) and on the swelling capacity of cultured astrocytes of newborn mice. If astrocytes were cultured on a substrate containing poly DL-ornithine, anti-AQP4 immunoreactivity was evenly and diffusely distributed. If, however, astrocytes were cultured in the presence of agrin-conditioned medium, we observed an increase in the intensity of AQP4-specific membrane-associated staining. Freeze-fracture studies revealed a clustering of orthogonal arrays of particles, representing a structural equivalent of AQP4, when exogenous agrin was present in the astrocyte cultures. Neuronal and non-neuronal agrin isoforms (agrin A0B0 and agrin A4B8, respectively) were able to induce membrane-associated AQP4 staining. Water transport capacity as well as the density of orthogonal arrays of intramembranous particles was increased in astrocytes cultured with the neuronal agrin isoform A4B8, but not with the endothelial and meningeal isoform A0B0. RT-PCR demonstrated that agrin A4B8 increased the level of the M23 splice variant of AQP4 and decreased the level of the M1 splice variant of AQP4. Implications for the regulation and maintenance of the blood-brain barrier including oedema formation under pathological conditions are discussed.
Collapse
Affiliation(s)
- Susan Noell
- Institute of Pathology, University of Tübingen, Liebermeisterstrasse 8, D-72076 Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
53
|
Karakaya S, Kipp M, Beyer C. Oestrogen regulates the expression and function of dopamine transporters in astrocytes of the nigrostriatal system. J Neuroendocrinol 2007; 19:682-90. [PMID: 17680883 DOI: 10.1111/j.1365-2826.2007.01575.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Dopamine is actively and specifically eliminated from the extracellular space by astrocytes and neurones through dopamine transporters (DAT) and, afterwards, either recycled into vesicles or metabolised. The availability of dopamine reflects a critical point in the regulation of dopamine activity within the nigrostriatal circuit under normal and pathological conditions. From previous studies, we know that oestrogen regulates the efficacy of dopaminergic neurones at the synaptic level and improves dopamine function during Parkinson's disease. Accordingly, we investigated the contribution of local astroglial for extracellular dopamine elimination and the impact of oestrogen on DAT expression and activity. Using neonatal striatal and midbrain astrocyte cultures, we could demonstrate that astrocytes possess a specific dopamine uptake machinery and express DAT at considerable levels. The application of 17beta-oestradiol decreased the expression of DAT by 80% and 60% in midbrain and striatal astroglia cultures, respectively. The unspecific dopamine transporters (OCT3, VMAT2) were not detected in astroglia. Functionally, oestrogen exposure inhibited the clearance of dopamine from the extracellular space by 45% and 35% compared to controls in midbrain and striatal astroglia, respectively. The effect on DAT expression and activity was completely antagonised by the oestrogen receptor antagonist ICI 182 780. In conclusion, our data suggest that the positive reinforcement of dopamine transmission under physiological conditions and the alleviative impact of oestrogen under pathological conditions may be the result of a decline in DAT expression and therefore delayed dopamine uptake by astroglia.
Collapse
Affiliation(s)
- S Karakaya
- Institute of Neuroanatomy, RWTH Aachen, 52074 Aachen, Germany
| | | | | |
Collapse
|
54
|
Quesada A, Romeo HE, Micevych P. Distribution and localization patterns of estrogen receptor-beta and insulin-like growth factor-1 receptors in neurons and glial cells of the female rat substantia nigra: localization of ERbeta and IGF-1R in substantia nigra. J Comp Neurol 2007; 503:198-208. [PMID: 17480015 PMCID: PMC2907103 DOI: 10.1002/cne.21358] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although several studies have focused on the neuroprotective effects of estrogen (E2) on stroke, there have been tantalizing reports on the potential neuroprotective role of E2 in degenerative neuronal diseases such as Alzheimer's and Parkinson's (PD). In animal models of PD, E2 protects the nigrostriatal dopaminergic (DA) system against neurotoxins. However, little is known about the cellular and molecular mechanism(s) involved by which E2 elicits its neuroprotective effects on the nigrostriatal DA system. A preferred mechanism for neuroprotection is the interaction of E2 with specific neuroprotective growth factors and receptors. One such neuroprotective factor/receptor system is insulin-like growth factor-1 (IGF-1). E2 neuroprotective effects in the substantia nigra (SN) DA system have been shown to be dependent on IGF-1. To determine whether E2 also interacts with the IGF-1 receptor (IGF-1R) and to determine the cellular localization of estrogen receptor (ER) and IGF-1R, we compared the distribution of ER and IGF-1R in the SN. Stereological measurements revealed that 40% of the subpopulation of tyrosine hydroxylase-immunoreactive (TH-ir) SN pars compacta (SNpc) DA neurons are immunoreactive for estrogen receptor-beta (ERbeta). No immunolabeling for ERalpha was observed. In situ hybridization and immunocytochemistry studies confirmed the expression of IGF-1R mRNA and revealed that almost all TH-ir SNpc DA neurons were immunoreactive for IGF-1R, respectively. Moreover, one-third of glial fibrillary acidic protein (GFAP-ir) cells in the SN were ERbeta-ir, and 67% of GFAP-ir cells expressed IGF-1R-ir. Therefore, the localization of ERbeta and IGF-1R on SNpc DA neurons and astrocytes suggests a modulatory role of E2 on IGF-1R, and this modulation may affect neuroprotection.
Collapse
Affiliation(s)
- Arnulfo Quesada
- Department of Neurobiology, Laboratory of Neuroendocrinology of the Brain Research Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095-1763, USA.
| | | | | |
Collapse
|
55
|
Kitano H, Young JM, Cheng J, Wang L, Hurn PD, Murphy SJ. Gender-specific response to isoflurane preconditioning in focal cerebral ischemia. J Cereb Blood Flow Metab 2007; 27:1377-86. [PMID: 17264860 PMCID: PMC2266686 DOI: 10.1038/sj.jcbfm.9600444] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Inhalation anesthetics are effective chemical preconditioning agents in experimental cerebral ischemia. However, previous work has been performed exclusively in male animals. We determined if there is a gender difference in ischemic outcome after isoflurane preconditioning (IsoPC), and if this sex-specific response is linked to differences in Akt phosphorylation or expression of neuronal inducible cell-death putative kinase (NIPK), a negative modulator of Akt activation. Young and middle-aged male and female mice were preconditioned for 4 h with air (sham PC) or 1.0% IsoPC and recovered for 24 h. Cortices were subdissected from preconditioned young male and female mice for measurement of Akt phosphorylation (Western blot) and NIPK mRNA (quantitative polymerase chain reaction). Additional cohorts underwent 2 h of reversible middle cerebral artery occlusion. Lastly, male and female Akt1(+/+) and Akt1(-/-) mice were studied to determine if gender differences in ischemic outcome after IsoPC is Akt1-dependent. Infarction volume was determined at 22 h reperfusion (2,3,5-triphenyltetrazolium chloride). As expected, IsoPC decreased ischemic damage as compared with sham PC in young and middle-aged male mice. In contrast, IsoPC markedly increased infarction in young female mice and had no effect in middle-aged female mice. Cortical phospho-Akt was increased by IsoPC versus sham PC only in male mice. No increase was observed in IsoPC female mice. NIPK mRNA was higher in female mice than in male mice regardless of preconditioning status. Male IsoPC neuroprotection was lost in Akt1-deficient male mice. We conclude that IsoPC is beneficial only in ischemic male brain and that sex differences in IsoPC are mediated through Akt activation and basal NIPK expression.
Collapse
Affiliation(s)
- Hideto Kitano
- Anesthesiology and Peri-Operative Medicine, Oregon Health and Science University, Portland, Oregon 97239-3098, USA
| | | | | | | | | | | |
Collapse
|
56
|
Kazi AA, Koos RD. Estrogen-induced activation of hypoxia-inducible factor-1alpha, vascular endothelial growth factor expression, and edema in the uterus are mediated by the phosphatidylinositol 3-kinase/Akt pathway. Endocrinology 2007; 148:2363-74. [PMID: 17272396 DOI: 10.1210/en.2006-1394] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Vascular endothelial growth factor (VEGF) plays an essential role in normal uterine physiology and function as well as endometrial cancer and other uterine disorders. Recently we showed that estrogen regulation of VEGF expression in the rat uterus involves rapid recruitment of both estrogen receptor (ER)-alpha and hypoxia-inducible factor (HIF)-1alpha to the VEGF promoter. Estrogen is known to stimulate both the MAPK and phosphatidylinositol 3-kinase (PI3K) pathways, which have been linked to the activation of both of these transcription factors. Therefore, the involvement of these pathways in estrogen-induced VEGF expression was investigated. Inhibitors of the MAPK (U0126) or PI3K pathways (wortmannin or LY294002) were administered ip to immature female rats 1 h before 17beta-estradiol (E(2)) treatment. E(2) activation of both pathways occurred and was completely inhibited by the appropriate antagonist. Only PI3K inhibitors, however, blocked E(2) stimulation of VEGF mRNA expression and E(2)-induced uterine edema. In vivo chromatin immunoprecipitation analysis showed that this was associated with a failure of both HIF-1alpha and ERalpha to bind to the VEGF promoter. To determine whether inhibiting the PI3K pathway affected ERalpha induction of other estrogen target genes, the expression of creatine kinase B and progesterone receptor A/B was also examined. The expression of each was also inhibited by wortmannin, as was ERalpha binding to the creatine kinase B promoter. In conclusion, although estrogen activates both the MAPK and PI3K pathways in the rat uterus, activation of HIF-1alpha and ERalpha, and therefore regulation of VEGF gene expression is dependent only on the PI3K/Akt pathway. Furthermore, activation of the PI3K pathway appears to be a common requirement for the expression of estrogen-induced genes. These findings not only shed light on estrogen action in normal target tissues but also have important implications for cancer biology because excessive PI3K, HIF-1alpha, and VEGF activity are common in estrogen-dependent tumors.
Collapse
Affiliation(s)
- Armina A Kazi
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1559, USA
| | | |
Collapse
|
57
|
Islamov RR, Valiullin VV, Murashov AK. Mechanisms of neuroprotective effect of estrogens associated with vascular endothelial growth factor expression. BIOL BULL+ 2007. [DOI: 10.1134/s1062359007020021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
58
|
Martínez-Cerdeño V, Noctor SC, Kriegstein AR. Estradiol stimulates progenitor cell division in the ventricular and subventricular zones of the embryonic neocortex. Eur J Neurosci 2007; 24:3475-88. [PMID: 17229096 DOI: 10.1111/j.1460-9568.2006.05239.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Two distinct populations of cerebral cortical progenitor cells that generate neurons during embryogenesis have been identified: radial glial cells and intermediate progenitor cells. Despite advances in our understanding of progenitor cell populations, we know relatively little about factors that regulate their proliferative behaviour. 17-beta-Estradiol (E2) is present in the adult and developing mammalian brain, and plays an important role in central nervous system processes such as neuronal differentiation, survival and plasticity. E2 also stimulates neurogenesis in the adult dentate gyrus. We examined the role of E2 during embryonic cortical neurogenesis through immunohistochemistry, in situ hybridization, functional enzyme assay, organotypic culture and in utero administration of estradiol-blocking agents in mice. We show that aromatase, the E2 synthesizing enzyme, is present in the embryonic neocortex, that estrogen receptor-alpha is present in progenitor cells during cortical neurogenesis, that in vitro E2 administration rapidly promotes proliferation, and that in utero blockade of estrogen receptors decreases proliferation of embryonic cortical progenitor cells. Furthermore, the E2 inhibitor alpha-fetoprotein is expressed at high levels by radial glial cells but at lower levels by intermediate progenitor cells, suggesting that E2 differentially influences the proliferation of these cortical progenitor cell types. These findings demonstrate a new functional role for E2 as a proliferative agent during critical stages of cerebral cortex development.
Collapse
Affiliation(s)
- Verónica Martínez-Cerdeño
- Department of Neurology and Program in Developmental and Stem Cell Biology, 513 Parnassus Avenue, HSW 1201, Box 0525, University of California San Francisco, San Francisco, California 94143, USA.
| | | | | |
Collapse
|
59
|
Abstract
Hormonal ligands for the nuclear receptor superfamily have at least two interacting mechanisms of action: 1) classical transcriptional regulation of target genes (genomic mechanisms); and 2) nongenomic actions that are initiated at the cell membrane, which could impact transcription. Although transcriptional mechanisms are increasingly well understood, membrane-initiated actions of these ligands are incompletely understood. Historically, this has led to a considerable divergence of thought in the molecular endocrine field. We have attempted to uncover principles of hormone action that are relevant to membrane-initiated actions of estrogens. There is evidence that the membrane-limited actions of hormones, particularly estrogens, involve the rapid activation of kinases and the release of calcium. Membrane actions of estrogens, which activate these rapid signaling cascades, can also potentiate nuclear transcription. These signaling cascades may occur in parallel or in series but subsequently converge at the level of modification of transcriptionally relevant molecules such as nuclear receptors and/or coactivators. In addition, other hormones or neurotransmitters may also activate cascades to crosstalk with estrogen receptor-mediated transcription. The idea of synergistic coupling between membrane-initiated and genomic actions of hormones fundamentally revises the paradigms of cell signaling in neuroendocrinology.
Collapse
Affiliation(s)
- Nandini Vasudevan
- Department of Biology, Pennsylvania State University, University Park, Pennsylvania 16802, USA.
| | | |
Collapse
|
60
|
Mendez P, Wandosell F, Garcia-Segura LM. Cross-talk between estrogen receptors and insulin-like growth factor-I receptor in the brain: cellular and molecular mechanisms. Front Neuroendocrinol 2006; 27:391-403. [PMID: 17049974 DOI: 10.1016/j.yfrne.2006.09.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Revised: 08/11/2006] [Accepted: 09/01/2006] [Indexed: 01/02/2023]
Abstract
Accumulating evidence suggests that insulin-like growth factor-I (IGF-I) and estradiol interact to regulate neural function. In this review, we focus on the cellular and molecular mechanisms involved in this interaction. The expression of estrogen receptors (ERs) and IGF-I receptor is cross-regulated in the central nervous system and many neurons and astrocytes coexpress both receptors. Furthermore, estradiol activates IGF-I receptor and its intracellular signaling. This effect may involve classical ERs since recent findings suggest that ERalpha may affect IGF-I actions in the brain by a direct interaction with some of the components of IGF-I signaling. In turn, IGF-I may regulate ER transcriptional activity in neuronal cells. In conclusion, ERs appear to be part of the signaling mechanism of IGF-I, and IGF-I receptor part of the mechanism of estradiol signaling in the nervous system.
Collapse
Affiliation(s)
- Pablo Mendez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), E-28002 Madrid, Spain
| | | | | |
Collapse
|
61
|
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
|
62
|
Guo RX, Wei LH, Tu Z, Sun PM, Wang JL, Zhao D, Li XP, Tang JM. 17 beta-estradiol activates PI3K/Akt signaling pathway by estrogen receptor (ER)-dependent and ER-independent mechanisms in endometrial cancer cells. J Steroid Biochem Mol Biol 2006; 99:9-18. [PMID: 16567092 DOI: 10.1016/j.jsbmb.2005.11.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Accepted: 11/30/2005] [Indexed: 11/26/2022]
Abstract
Cellular response to estrogen is mediated both by estrogen receptor (ER) binding to estrogen response element (ERE) and by non-nuclear actions like activation of signal transducing pathways. The main aims are to study if PI3K/Akt signaling pathway can be activated by 17beta-estradiol (E2) via non-nuclear action and to investigate the relationship of the action of E2 and ER in endometrial cancer cells expressing with different status of ER. The levels of phosphorylated Akt (Ser473) (P-Akt) and total Akt were examined by western blot and Akt kinase activity was measured in cells after stimulation with 1 microM E2 at different time points. Inhibitory role of LY294002 on activation of Akt induced by E2 and its estrogen antagonist, ICI182780 were also tested. P-Akt/Akt was used as a measure of activation of Akt. We found that maximum P-Akt/Akt and Akt kinase activity took place at 30 min in Ishikawa cells and 15 min in HEC-1A cells and the activation persisted for at least 2 h after stimulation with 1 microM E2. The activation of Akt elicited gradually with increasing doses of E2. PI3K inhibitor, LY294002, stopped the activating Akt in a dose-dependent manner and 50 microM LY294002 completely blocked the activation of Akt induced by E2. ICI182780 could block the activation of PI3K/Akt in ER-positive Ishikawa cells but not in HEC-1A cells with poor-expressed ER. This study demonstrated that E2 is able to promptly activate PI3K/Akt signal pathway in Ishikawa cells in an ER-dependent manner and ER-independent in HEC-1A cells. Blockage of PI3K/Akt cascade may become a potential and effective way to control endometrial carcinoma, especially in ER-negative cancers, which show no response to endocrinal therapy.
Collapse
Affiliation(s)
- Rui-Xia Guo
- Department of Gynecology, Peking University, People's Hospital, No. 11, XiZhiMen South Street, Xi Cheng District, Beijing 100044, China
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Kalkman HO. The role of the phosphatidylinositide 3-kinase–protein kinase B pathway in schizophrenia. Pharmacol Ther 2006; 110:117-34. [PMID: 16434104 DOI: 10.1016/j.pharmthera.2005.10.014] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Accepted: 10/26/2005] [Indexed: 01/05/2023]
Abstract
Neuroanatomical studies of brains from schizophrenic patients report evidence for neuronal dystrophy, while in genetic studies in schizophrenia there is evidence for mutations in growth factors and the downstream enzymes phosphatidylinositide 3-kinase (PI3K) and protein kinase B (PKB). Since the PI3K-PKB pathway is involved in cellular growth and proliferation, reduced activity of this cascade in schizophrenia could at least partly explain the neuronal dystrophy. Risk factors for schizophrenia, such as corticosteroids and cannabis, suppress the activity of the PI3K-PKB pathway. Conversely, estrogen and vitamin D, 2 factors with a moderate protective activity in schizophrenia, electroconvulsive shock therapy, and chronic treatment with antipsychotic compounds stimulate the pathway. Reduced activity of the PI3K-PKB pathway makes the brain more susceptible to virus infections, anoxia, and obstetric complications (recognized risk factors for schizophrenia), whereas a diminution of growth factor levels towards the end of puberty could contribute to an increase in schizophrenia symptoms observed around that time. On the other hand, constitutive (over)activation of the PI3K-PKB pathway increases cancer risk. Consequently, the presumed hypoactivity of the PI3K-PKB cascade might provide a partial explanation for the remarkable epidemiological finding of a reduced cancer rate in schizophrenic patients. Recognition of the role of a dysfunctional PI3K-PKB pathway in schizophrenia might help in the discovery of hitherto undetected causative gene mutations and could also lead to novel therapeutic approaches. However, a major challenge that remains to be solved is how the PI3K-PKB pathway can be activated without increasing the risk of cancer.
Collapse
Affiliation(s)
- Hans O Kalkman
- Neuroscience Research, Novartis Institutes of Biomedical Research Basel, Building WSJ-360.4.05, Novartis Pharma AG, CH4002 Basel, Switzerland.
| |
Collapse
|
64
|
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
|
65
|
Toyofuku A, Hara T, Taguchi T, Katsura Y, Ohama K, Kudo Y. Cyclic and characteristic expression of phosphorylated Akt in human endometrium and decidual cells in vivo and in vitro. Hum Reprod 2005; 21:1122-8. [PMID: 16373405 DOI: 10.1093/humrep/dei454] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Akt is activated by phosphorylation and plays an important role in cell survival and maintenance of structure. METHODS We investigated whether phosphorylated Akt was characteristically expressed in human endometrium in vivo and whether insulin-like growth factor-I (IGF-I) can activate Akt using cultured decidualized human stromal cells in vitro, using immunohistochemistry and Western blotting analysis. RESULTS The levels of phosphorylated Akt protein increased markedly in the decidual tissues from ectopic pregnancy. The expression of phosphorylated Akt protein in stromal cells increased with the decidualization. The decidual cells showed strong cytoplasmic staining for phosphorylated Akt. However, cultured decidualized human stromal cells diminished phosphorylated Akt expression compared to control cells. IGF-I administration to decidualized human stromal cells significantly recovered pAkt expression. The effect of IGF-I on decidualized human stromal cells was blocked by an inhibitor of phosphatidylinositol-3 kinase (PI3K) (LY294,002). These results suggest that IGF-I may activate Akt via PI3K in human endometrium and decidua. The expression of phosphorylated Akt in stromal cells was only detected in the functional layer, where tissue remodelling occurs during menstruation or implantation. CONCLUSIONS Akt activation may be involved in cell survival and extracellular matrix remodelling in human endometrium and decidua.
Collapse
Affiliation(s)
- Aya Toyofuku
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | |
Collapse
|
66
|
Sun RQ, Tu YJ, Yan JY, Willis WD. Activation of protein kinase B/Akt signaling pathway contributes to mechanical hypersensitivity induced by capsaicin. Pain 2005; 120:86-96. [PMID: 16360265 DOI: 10.1016/j.pain.2005.10.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2005] [Revised: 09/09/2005] [Accepted: 10/19/2005] [Indexed: 11/20/2022]
Abstract
We investigated the involvement of the protein kinase B/Akt (PKB/Akt) signaling pathway in the mechanical hypersensitivity induced in rats by capsaicin. Intradermal injection of capsaicin results in activation of PKB/Akt in the lumbar spinal cord, most prominently in the dorsal horn, starting by 5 min after capsaicin injection and lasting at least 1h. The activated PKB/Akt in the spinal cord is in neurons, since phospho-PKB/Akt (p-PKB/Akt) colocalizes with the neuronal marker, neuronal-specific nuclear protein (NeuN). The mechanical hypersensitivity is shown by the enhanced paw withdrawal frequency to applications of von Frey filaments with different bending forces (30, 100, 200 mN) on the rat paw. Pre-treatment with several different PKB/Akt inhibitors, including SH-6, Akt inhibitor IV, and Akt inhibitor V, blocked the mechanical hypersensitivity induced by intradermal injection of capsaicin, a measure of spinal cord central sensitization. Two structurally unrelated phosphoinositide 3-Kinase (PI3K, upstream of PKB/Akt) inhibitors, Wortmannin and LY294002, also prevented the mechanical hypersensitivity induced by intradermal injection of capsaicin. Furthermore, post-treatment with the PI3K inhibitor, Wortmannin, or PKB/Akt inhibitors, such as NL-71-101, SH-6, Akt inhibitor IV, and inhibitor V significantly reduced the established mechanical hypersensitivity induced by capsaicin. The PKB/Akt signaling pathway in the spinal cord is therefore involved in pain hypersensitivity.
Collapse
Affiliation(s)
- Rui-Qing Sun
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1069, USA
| | | | | | | |
Collapse
|
67
|
Mhyre AJ, Dorsa DM. Estrogen activates rapid signaling in the brain: role of estrogen receptor alpha and estrogen receptor beta in neurons and glia. Neuroscience 2005; 138:851-8. [PMID: 16343790 DOI: 10.1016/j.neuroscience.2005.10.019] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Revised: 09/14/2005] [Accepted: 10/10/2005] [Indexed: 10/25/2022]
Abstract
The aging process is known to coincide with a decline in circulating sex hormone levels in both men and women. Due to an increase in the average lifespan, a growing number of post-menopausal women are now receiving hormone therapy for extended periods of time. Recent findings of the Women's Health Initiative, however, have called into question the benefits of long-term hormone therapy for treating symptoms of menopause. The results of this study are still being evaluated, but it is clear that a better understanding of the molecular effects of estradiol is needed in order to develop new estrogenic compounds that activate specific mechanisms but lack adverse side effects. Traditionally, the effects of estradiol treatment have been ascribed to changes in gene expression, namely transcription at estrogen response elements. This review focuses on emerging information that estradiol can also activate a repertoire of membrane-initiated signaling pathways and that these rapid signaling events lead to functional changes at the cellular level. The various types of cells in the brain can respond differently to estradiol treatment based on the signaling properties of the cell, as well as which receptor, estrogen receptor alpha and/or estrogen receptor beta, is expressed. Taken together, these findings suggest that the estradiol-induced activation of membrane-initiated signaling pathways occurs in a cell-type specific manner and can differentially influence how the cells respond to various insults.
Collapse
Affiliation(s)
- A J Mhyre
- Department of Pharmacology, University of Washington School of Medicine, Seattle, 98195, USA
| | | |
Collapse
|
68
|
Chen JQ, Contreras RG, Wang R, Fernandez SV, Shoshani L, Russo IH, Cereijido M, Russo J. Sodium/potasium ATPase (Na+, K+-ATPase) and ouabain/related cardiac glycosides: a new paradigm for development of anti- breast cancer drugs? Breast Cancer Res Treat 2005; 96:1-15. [PMID: 16322895 DOI: 10.1007/s10549-005-9053-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Accepted: 08/11/2005] [Indexed: 11/30/2022]
Abstract
Prolonged exposure to 17beta-estradiol (E2) is a key etiological factor for human breast cancer. The biological effects and carcinogenic effects of E2 are mediated via estrogen receptors (ERs), ERalpha and ERbeta. Anti-estrogens, e.g. tamoxifen, and aromatase inhibitors have been used to treat ER-positive breast cancer. While anti-estrogen therapy is initially successful, a major problem is that most tumors develop resistance and the disease ultimately progresses, pointing to the need of developing alternative drugs targeting to other critical targets in breast cancer cells. We have identified that Na+, K+-ATPase, a plasma membrane ion pump, has unique/valuable properties that could be used as a potentially important target for breast cancer treatment: (a) it is a key player of cell adhesion and is involved in cancer progression; (b) it serves as a versatile signal transducer and is a target for a number of hormones including estrogens and (d) its aberrant expression and activity are implicated in the development and progression of breast cancer. There are several lines of evidence indicating that ouabain and related digitalis (the potent inhibitors of Na+, K+-ATPase) possess potent anti-breast cancer activity. While it is not clear how the suggested anti-cancer activity of these drugs work, several observations point to ouabain and digitalis as being potential ER antagonists. We critically reviewed many lines of evidence and postulated a novel concept that Na+, K+-ATPase in combination with ERs could be important targets of anti-breast cancer drugs. Modulators, e.g. ouabain and related digitalis could be useful to develop valuable anti-breast cancer drugs as both Na+, K+-ATPase inhibitors and ER antagonists.
Collapse
Affiliation(s)
- Jin-Qiang Chen
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Pawlak J, Brito V, Küppers E, Beyer C. Regulation of glutamate transporter GLAST and GLT-1 expression in astrocytes by estrogen. ACTA ACUST UNITED AC 2005; 138:1-7. [PMID: 15896872 DOI: 10.1016/j.molbrainres.2004.10.043] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Revised: 10/11/2004] [Accepted: 10/24/2004] [Indexed: 11/18/2022]
Abstract
Estrogen influences neuronal development and a broad spectrum of neural functions. In addition, several lines of evidence suggest a role as neuroprotective factor for estrogen in the CNS. Neuroprotection can result from direct estrogen-neuron interactions or be mediated indirectly involving the regulation of physiological properties of nonneuronal cells, such as astrocytes and microglia. Increased l-glutamate levels are associated with neurotoxic and neurodegenerative processes in the brain. Thus, the removal of l-glutamate from the extracellular space by astrocytes through the astroglial glutamate transporters GLT-1 and GLAST appears essential for maintaining a homeostatic milieu for neighboring neurons. We have therefore studied the influence of 17beta-estradiol on l-glutamate metabolism in cultured astrocytes from the neonate mouse midbrain using quantitative RT-PCR and Western blotting for both transporters as well as functional l-glutamate uptake studies. The administration of estrogen significantly increased the expression of GLT-1 and GLAST on the mRNA and protein level. Likewise, specific l-glutamate uptake by astrocytes was elevated after estrogen exposure and mimicked by dbcAMP stimulation. Induction of transporter expression and l-glutamate uptake were sensitive to ICI 182,780 treatment suggesting estrogen action through nuclear estrogen receptors. These findings indicate that estrogen can prevent l-glutamate-related cell death by decreasing extracellular l-glutamate levels through an increased l-glutamate uptake capacity by astrocytes.
Collapse
Affiliation(s)
- Justyna Pawlak
- Anatomisches Institut, Universität Tübingen, Osterbergstr. 3, 72074 Tübingen, Germany
| | | | | | | |
Collapse
|
70
|
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
|
71
|
Van de Sande T, Roskams T, Lerut E, Joniau S, Van Poppel H, Verhoeven G, Swinnen JV. High-level expression of fatty acid synthase in human prostate cancer tissues is linked to activation and nuclear localization of Akt/PKB. J Pathol 2005; 206:214-9. [PMID: 15880754 DOI: 10.1002/path.1760] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many human epithelial cancers, particularly those with a poor prognosis, express high levels of fatty acid synthase (FAS), a key metabolic enzyme linked to the synthesis of membrane phospholipids in cancer cells. In view of the recent finding that in the human prostate cancer cell line LNCaP, overexpression of FAS can be largely attributed to constitutive activation of the phosphatidylinositol-3 (PI3) kinase/Akt kinase pathway, the activation status of the Akt pathway, and whether this activation coincides with increased FAS expression, was examined in clinical prostate cancer tissues. Using well-preserved frozen prostatic needle biopsies and a sensitive Envision detection technique, S473-phosphorylated Akt (pAkt) was found in 11/23 low-grade prostatic intraepithelial neoplasia (PIN) lesions, in all (36/36) high-grade PINs, and in all (86/86) invasive carcinomas. Non-neoplastic tissues were negative. Interestingly, in low-grade PINs and low-grade carcinomas, pAkt was mainly cytoplasmic or membrane-bound and was associated with moderate elevation of FAS expression. In 24/36 high-grade PINs and 82/88 invasive carcinomas, pAkt was found at least partly in the nucleus. Greater nuclear pAkt staining, and higher FAS expression, correlated with a higher Gleason score. In the light of previous findings that pAkt plays a causative role in the overexpression of FAS in cancer cells in culture, these data strongly suggest that high-level expression of FAS in prostate cancer tissues is linked to phosphorylation and nuclear accumulation of Akt.
Collapse
Affiliation(s)
- Tine Van de Sande
- Laboratory for Experimental Medicine and Endocrinology, University of Leuven, B-3000 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
72
|
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
|
73
|
Geffroy N, Guédin A, Dacquet C, Lefebvre P. Cell cycle regulation of breast cancer cells through estrogen-induced activities of ERK and Akt protein kinases. Mol Cell Endocrinol 2005; 237:11-23. [PMID: 15925090 DOI: 10.1016/j.mce.2005.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Revised: 04/05/2005] [Accepted: 04/06/2005] [Indexed: 11/26/2022]
Abstract
The proliferative effect of estrogens on breast cancer cell (BCC) is mainly mediated through estrogen receptors (ER). Non-transcriptional effects of estrogens, exerted through activation of several protein kinases, may also contribute to BCC proliferation. However, the relative contribution of these two responses to BCC proliferation is not known. We characterized a novel estrogenic receptor ligand which possess Akt and ERK activating properties distinct from that of 17beta-estradiol. Early and delayed waves of activation of these kinases were detected upon estrogenic challenge of BCC, but only molecules able to promote a significant, delayed activation of ERK-induced BCC proliferation. Estrogen-induced cell cycle progression was not sensitive to the inhibition of ERK-regulating kinases MEK1 and 2. ERalpha was found to be necessary, but not sufficient for kinases activation. Thus, estrogens elicit a distinct pattern of early and delayed activation of ERK and Akt, and early protein kinase activation is probably not involved in BCC proliferation. Structural variations in the estrogen molecule may confer novel biological properties unrelated to estrogen-dependent transcriptional activation.
Collapse
Affiliation(s)
- Nancy Geffroy
- INSERM U 459, Faculté de Médecine Henri Warembourg, 1 place de Verdun, 59045 Lille Cedex, France
| | | | | | | |
Collapse
|
74
|
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
|
75
|
Garcia-Ovejero D, Azcoitia I, Doncarlos LL, Melcangi RC, Garcia-Segura LM. Glia-neuron crosstalk in the neuroprotective mechanisms of sex steroid hormones. ACTA ACUST UNITED AC 2005; 48:273-86. [PMID: 15850667 DOI: 10.1016/j.brainresrev.2004.12.018] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2004] [Accepted: 12/09/2004] [Indexed: 11/22/2022]
Abstract
Proteins involved in the intramitochondrial trafficking of cholesterol, the first step in steroidogenesis, such as the steroidogenic acute regulatory protein (StAR) and the peripheral-type benzodiazepine receptor (PBR), are upregulated in the nervous system after injury. Accordingly, a local increase in the levels of steroids, such as pregnenolone and progesterone, is observed following traumatic injury in the brain and spinal cord. The expression and activity of aromatase, the enzyme that synthesizes estradiol, is also increased in injured brain areas and its inhibition results in an increased neurodegeneration. These findings suggest that an increase in steroidogenesis is part of an overall mechanism used by the nervous tissue to cope with neurodegenerative conditions. Neural steroidogenesis is the result of a coordinated interaction of neurons and glia. For example, after neural injury, there is an upregulation of StAR in neurons and of PBR in microglia and astroglia. Aromatase is expressed in neurons under basal conditions and is upregulated in reactive astrocytes after injury. Some of the steroids produced by glia are neuroprotective. Progesterone and progesterone derivatives produced by Schwann cells, promote myelin formation and the remyelination and regeneration of injured nerves. In the central nervous system, the steroids produced by glia regulate synaptic function, affect anxiety, cognition, sleep and behavior, and exert neuroprotective and reparative roles. In addition, glial cells are targets for steroids and mediate some of the effects of these molecules on neurons, including the regulation of survival and regeneration.
Collapse
|
76
|
Temple JL, Laing E, Sunder A, Wray S. Direct action of estradiol on gonadotropin-releasing hormone-1 neuronal activity via a transcription-dependent mechanism. J Neurosci 2004; 24:6326-33. [PMID: 15254088 PMCID: PMC6729551 DOI: 10.1523/jneurosci.1006-04.2004] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2004] [Revised: 05/21/2004] [Accepted: 06/02/2004] [Indexed: 11/21/2022] Open
Abstract
Pulsatile secretion of gonadotropin-releasing hormone-1 (GnRH-1) is essential for reproduction. GnRH-1 induces gonadotropin release and is regulated by 17beta-estradiol (E2). Although a subpopulation of GnRH-1 neurons expresses estrogen receptor (ER) beta, it is unclear whether E2 acts directly on GnRH-1 neurons or indirectly through interneuronal connections. To test the hypothesis that E2 acts directly on GnRH-1 neurons to regulate neuronal activity, we used calcium imaging to monitor intracellular calcium oscillations in GnRH-1 neurons maintained in nasal explants. TTX was used to minimize synaptic input from other cells. Consistent with previous studies, TTX reduced the activity of individual GnRH-1 neurons to a basal level, while the population of cells maintained synchronized calcium oscillations. Exposure of GnRH-1 cells to TTX plus E2 increased the number of calcium peaks/cell, percentage of cells with > or =10 peaks, mean peak amplitude, and percentage of cells that contributed to each calcium pulse in explants maintained in vitro for 7 d (7 div) compared with TTX alone. These effects were induced within 30 min and were not mimicked by 17alpha-estradiol, E2 conjugated to BSA (which does not cross the plasma membrane), or seen at 21 div, when the percentage of GnRH-1 cells expressing ERbeta transcripts declines. In addition, these effects were inhibited by the ER antagonist ICI 182,780 and prevented by inhibition of gene transcription. These data suggest that, via ERbeta, E2 can rapidly act as a hormone-activated transcription complex and are the first to show that E2 directly increases GnRH-1 neuronal activity and synchronization.
Collapse
Affiliation(s)
- Jennifer L Temple
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
77
|
Cardona-Gomez P, Perez M, Avila J, Garcia-Segura LM, Wandosell F. Estradiol inhibits GSK3 and regulates interaction of estrogen receptors, GSK3, and beta-catenin in the hippocampus. Mol Cell Neurosci 2004; 25:363-73. [PMID: 15033165 DOI: 10.1016/j.mcn.2003.10.008] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2003] [Revised: 09/24/2003] [Accepted: 10/07/2003] [Indexed: 12/25/2022] Open
Abstract
Estrogens regulate a wide set of neuronal functions such as gene expression, survival and differentiation in a manner not very different from that exerted by neurotrophins or by growth factors. The best-studied hormonal action is the transcriptional activation mediated by estrogen receptors. However, the direct effects of estrogen on growth factor signaling have not been well clarified. The present data show that estradiol, in vivo, induces a transient activation of GSK3 in the adult female rat hippocampus, followed by a more sustained inhibition, as inferred from phosphorylation levels of Tau. Similar data was obtained from cultured hippocampal neurons when treated with the hormone. The transient activation was confirmed by direct measure of GSK3 kinase activity. In addition, our results show a novel complex of estrogen receptor alpha, GSK3, and beta-catenin. The presence of the hormone removes beta-catenin from this complex. There is a second complex, also affected by estradiol, in which Tau is associated with GSK3, beta-catenin, and elements of the PI3 kinase complex. Considering the role of GSK3 in neurodegeneration, our data suggest that part of the neuroprotective effects of estrogen may be due to the control of GSK3.
Collapse
Affiliation(s)
- P Cardona-Gomez
- Centro de Biología Molecular Severo Ochoa CSIC-Universidad Autónoma de Madrid, Madrid 28049, Spain
| | | | | | | | | |
Collapse
|
78
|
Murashov AK, Islamov RR, McMurray RJ, Pak ES, Weidner DA. Estrogen increases retrograde labeling of motoneurons: evidence of a nongenomic mechanism. Am J Physiol Cell Physiol 2004; 287:C320-6. [PMID: 15044155 DOI: 10.1152/ajpcell.00542.2003] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Estrogen has a variety of neurotrophic effects mediated via different signaling cascades, including ERK and phosphatidylinositol 3-kinase (PI3K) pathways. In this study, we investigated effects of estrogen and inhibitors for ERK and PI3K applied directly onto the cut sciatic nerve on retrograde labeling of lumbar motoneurons. A mix of retrograde tracer (Fluorogold) and 17beta-estradiol, in combination with an antagonist for estrogen receptors ICI 182,780, an inhibitor of ERK1/2 pathway (U0126), an inhibitor of PI3K (LY-294002), or a protein synthesis inhibitor (cycloheximide), was applied to the proximal stump of the transected sciatic nerve for 24 h. Coapplication of Fluorogold with 17beta-estradiol produced a significant increase in the number of retrograde-labeled lumbar motoneurons, compared with Fluorogold alone. Estrogen potentiation of retrograde labeling was inhibited by application of ICI 182,780, U0126, LY-294002, and cycloheximide. Immunohistochemical analysis of the sciatic nerve, 24 h following crush injury, revealed accumulation of phospho-ERK in regenerating nerve fibers. The data suggest a role for estrogen, ERK, PI3K, and protein synthesis in the uptake and retrograde transport of Fluorogold. We propose that estrogen action in peripheral nerve fibers is mediated via the ERK and PI3K signaling pathways and is reliant on local protein synthesis.
Collapse
Affiliation(s)
- Alexander K Murashov
- Department of Physiology, Brody School of Medicine, East Carolina University, Brody Bldg. #6N-98, 600 Moye Blvd., Greenville, NC 27858, USA.
| | | | | | | | | |
Collapse
|
79
|
Abstract
The ovarian hormone 17beta-estradiol (E2) is neuroprotective in animal models of neurodegenerative diseases. Some studies suggest that the neuroprotective effects of 17beta-estradiol are a consequence of its antioxidant activity that depend on the hydroxyl group in the C3 position of the A ring. As in other tissues, 17beta-estradiol is metabolized in the brain to 2-hydroxyestradiol (2OHE2) and 2-methoxyestradiol (2MEOHE2). These two molecules present the hydroxyl group in the A ring and have a higher antioxidant activity than 17beta-estradiol. To test the hypothesis that conversion to 2-hydroxyestradiol and 2-methoxyestradiol may mediate neuroprotective actions of 17beta-estradiol in vivo, we have assessed whether these molecules protect hilar hippocampal neurons from kainic acid toxicity. Ovariectomized Wistar rats received an i.p. injection of 1, 10 or 100 microg 17beta-estradiol, 2-hydroxyestradiol or 2-methoxyestradiol followed by an i.p. injection of kainic acid (7 mg/kg) or vehicle. Treatment with kainic acid resulted in a significant loss of hilar neurons. Only the highest dose tested of 17beta-estradiol (100 microg/rat) prevented kainic acid-induced neuronal loss. 2-Hydroxyestradiol and 2-methoxyestradiol did not protect hilar neurons from kainic acid, suggesting that the mechanism of neuroprotection by 17beta-estradiol in vivo is not mediated by its metabolism to catecholestrogens or methoxycatecholestrogens. Furthermore, 2-methoxyestradiol (100 microg/rat), by itself, resulted in a significant neuronal loss in the hilus that was detected 96 h after the treatment with the steroid. This finding suggests that endogenous metabolism of 17beta-estradiol to 2-methoxyestradiol may counterbalance the neuroprotective effects of the hormone.
Collapse
Affiliation(s)
- Ofir Picazo
- Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón Col. Sto. Tomás 11340, México D.F., Mexico
| | | | | |
Collapse
|
80
|
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
|
81
|
Rider V, Thomson E, Seifert C. Transit of rat uterine stromal cells through G1 phase of the cell cycle requires temporal and cell-specific hormone-dependent changes on cell cycle regulators. Endocrinology 2003; 144:5450-8. [PMID: 12960012 DOI: 10.1210/en.2003-0890] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Progesterone pretreatment increases the number of synchronously proliferating stromal cells in the ovariectomized rat uterus, but estrogen is necessary to stimulate reentry into the cell cycle. To investigate the mechanisms underlying differential hormone actions, sexually mature ovariectomized rats were injected with progesterone (2 mg) for three consecutive days. Estradiol 17-beta (0.6 microg) was administered to initiate cell proliferation. Uterine samples were collected at timed intervals. Cell entry into DNA replication was monitored by injecting 5-bromo-2'-deoxyuridine (1 mg/100 g body weight) 2 h before necropsy. Demicolchicine (400 microg) was injected 30 min before necropsy to assess transit into M phase. Temporal progress through G1 was determined by spatial changes in cyclin D1/D3 proteins. Total cyclin D1/D3 protein and mRNA was measured by Western and Northern blotting. Estrogen increased the number of 5-bromo-2'-deoxyuridine-positive stromal cells (P < 0.05), compared with the number in rats treated with progesterone alone. An increase (P < 0.05) in the number of M-phase cells occurred at 12 h post estrogen. There was no evidence for epithelial cell proliferation in response to steroid treatments. Cyclin D1/D3 mRNA was expressed in the uteri of ovariectomized and hormone treated rats. The D-type cyclin proteins, however, were not evident in stromal cells without estrogen treatment. Progesterone pretreatment inhibited estrogen-dependent epithelial cell proliferation while redirecting D-type cyclin expression to the uterine stroma. Stromal cell transit through G1 required nongenomic steroid-dependent action on signal transduction pathways that control the nuclear localization and cell type-specific expression of the D-type cyclin proteins.
Collapse
Affiliation(s)
- Virginia Rider
- Department of Biology, Pttsburg State University, Pittsburg, Kansas 66762, USA.
| | | | | |
Collapse
|
82
|
Xu Y, Traystman RJ, Hurn PD, Wang MM. Neurite-localized estrogen receptor-alpha mediates rapid signaling by estrogen. J Neurosci Res 2003; 74:1-11. [PMID: 13130501 DOI: 10.1002/jnr.10725] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Classically, estrogen acts on cells by directly activating gene transcription driven by ligand-bound nuclear estrogen receptors (ER). Accumulating evidence demonstrates that estrogen acts on neurons by utilizing diverse molecular mechanisms, including rapid signaling by proteins localized to the plasma membrane. Recent studies showing that ERalpha localizes to axons and dendrites of hippocampal neurons suggest that nonnuclear stores of the receptor may transduce estrogen signaling. Here, we have studied the subcellular localization, dynamic regulation, and function of ERalpha in mouse cortical neurons. Estrogen-stimulated mouse cortical neurons activate both estrogen response element (ERE) stimulated transcription and rapid activation of p44/42 mitogen-activated protein kinases (MAPK). We demonstrate that green fluorescent protein (GFP)-tagged ERalpha localizes to neurites in cultured cortical neurons and that the expression within neurites can be down-regulated by estrogen or up-regulated by antiestrogen administered during synthesis. Neurite ERalpha appears to be directed to neurites directly from its site of translation and not from nuclear stores. By using confocal microscopy, we show that ERalpha within neurites stimulates local activation of p44/42 MAP kinases in response to estrogen. We conclude that hormonal status alters subcellular ERalpha targeting in cortical neurons and that neurite-expressed ERalpha is important in the activation of local MAPK signaling.
Collapse
Affiliation(s)
- Yun Xu
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | | | | | | |
Collapse
|
83
|
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
|
84
|
Estrogen receptor beta mediates rapid estrogen actions on gonadotropin-releasing hormone neurons in vivo. J Neurosci 2003. [PMID: 12843281 DOI: 10.1523/jneurosci.23-13-05771.2003] [Citation(s) in RCA: 148] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The gonadal steroid estrogen exerts an important modulatory influence on the activity of multiple neuronal networks. In addition to classical genomic mechanisms of action, estrogen also exerts poorly understood rapid, nongenomic effects on neurons. To examine whether estrogen may exert rapid actions on intracellular signaling within gonadotropin-releasing hormone (GnRH) neurons in vivo,we examined the phosphorylation status of cAMP response element-binding protein (CREB) in these cells after the administration of 17-beta-estradiol to ovariectomized (OVX) mice. The percentage of GnRH neurons expressing phosphorylated CREB was increased more than sixfold (p < 0.05) in a time- and dose-dependent manner by estrogen, with the increase first observed 15 min after estrogen administration. A series of in vitro studies demonstrated that estrogen acted directly on native GnRH neurons to phosphorylate CREB, but that estrogen conjugated to bovine serum albumin was without effect. The role of classical estrogen receptors (ERs) was evaluated using ER knock-out mice in vivo. The effect of estrogen on CREB phosphorylation in GnRH neurons was normal in ERalpha knock-out mice but completely absent in ERbeta knock-out mice. Finally, studies in intact female mice revealed levels of CREB phosphorylation within GnRH neurons that were equivalent to those of estrogen-treated OVX mice. These observations demonstrate that ERbeta mediates the rapid, direct effects of estrogen on the GnRH neuronal phenotype, and that these actions persist under physiological conditions. They also provide the first evidence for a role of ERbeta in nongenomic estrogen signaling within the brain in vivo.
Collapse
|
85
|
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
|
86
|
Marino M, Acconcia F, Trentalance A. Biphasic estradiol-induced AKT phosphorylation is modulated by PTEN via MAP kinase in HepG2 cells. Mol Biol Cell 2003; 14:2583-91. [PMID: 12808053 PMCID: PMC194905 DOI: 10.1091/mbc.e02-09-0621] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We reported previously in HepG2 cells that estradiol induces cell cycle progression throughout the G1-S transition by the parallel stimulation of both PKC-alpha and ERK signaling molecules. The analysis of the cyclin D1 gene expression showed that only the MAP kinase pathway was involved. Here, the presence of rapid/nongenomic, estradiol-regulated, PI3K/AKT signal transduction pathway, its modulation by the levels of the tumor suppressor PTEN, its cross-talk with the ERK pathway, and its involvement in DNA synthesis and cyclin D1 gene promoter activity have all been studied in HepG2 cells. 17beta-Estradiol induced the rapid and biphasic phosphorylation of AKT. These phosphorylations were independent of each other, being the first wave of activation independent of the estrogen receptor (ER), whereas the second was dependent on ER. Both activations were dependent on PI3K activity; furthermore, the ERK pathway modulated AKT phosphorylation by acting on the PTEN levels. The results showed that the PI3K pathway, as well as ER, were strongly involved in both G1-S progression and cyclin D1 promoter activity by acting on its proximal region (-254 base pairs). These data indicate that in HepG2 cells, different rapid/nongenomic estradiol-induced signal transduction pathways modulate the multiple steps of G1-S phase transition.
Collapse
Affiliation(s)
- Maria Marino
- Dipartimento di Biologia, Università Roma Tre, V. le G. Marconi, 446, Italy.
| | | | | |
Collapse
|
87
|
Estrogen stimulates postsynaptic density-95 rapid protein synthesis via the Akt/protein kinase B pathway. J Neurosci 2003. [PMID: 12657692 DOI: 10.1523/jneurosci.23-06-02333.2003] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Estrogens induce synaptogenesis in the CA1 region of the dorsal hippocampus during the estrous cycle of the female rat. Functional consequences of such estrogen-mediated synaptogenesis include cyclic changes in neurotransmission and memory. At the molecular level, estrogen stimulates the rapid activation of specific signal transduction pathways, and of particular interest is the activation of Akt (protein kinase B), a key signal transduction intermediate that initiates protein translation by alleviating the downstream translational repression of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1). Using a well established in vitro model system of differentiated NG108-15 neurons to investigate such rapid signaling effects of estrogen, we show that estrogen stimulates the phosphorylation of Akt, an indication of kinase activation, as well as the phosphorylation of 4E-BP1. In turn, the activation of these signaling intermediates suggests a non-genomic mechanism by which estrogen might likewise lead to protein translation of dendrite-localized mRNA transcripts in the hippocampus in vivo. We therefore considered the translation of the dendritic spine scaffolding protein postsynaptic density-95 (PSD-95). Although estrogen does not stimulate a rapid increase in PSD-95 mRNA levels in NG108-15 neurons, we show here that estrogen does however stimulate a rapid increase in PSD-95 new protein synthesis in vitro and that this new protein synthesis is Akt dependent. These results demonstrate an essential role for Akt in estrogen-stimulated dendritic spine protein expression, describe for the first time a signal transduction pathway in PSD-95 expression, and delineate a novel, molecular mechanism by which ovarian hormones might translationally regulate synaptogenesis via activating protein synthesis for dendritic function.
Collapse
|
88
|
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
|
89
|
Islamov RR, Hendricks WA, Katwa LC, McMurray RJ, Pak ES, Spanier NS, Murashov AK. Effect of 17 beta-estradiol on gene expression in lumbar spinal cord following sciatic nerve crush injury in ovariectomized mice. Brain Res 2003; 966:65-75. [PMID: 12646309 DOI: 10.1016/s0006-8993(02)04191-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Previously, we observed that estrogen treatment enhances regeneration of the sciatic nerve after crush injury [Brain Res. 943 (2002) 283]. In this research, we studied expression of estrogen receptors and effects of estrogen on gene expression in the lumbar spinal cord, following sciatic nerve crush injury. Using the Atlas Mouse 1.2 Array, changes in the expression of 267 of 1176 genes were registered 4 days after nerve injury. Those genes that exhibited a change in signal intensity ratios of 2-fold or greater were selected as up-regulated (42) or down-regulated (21). In estrogen treated mice, we have observed up-regulation of the genes known to control apoptosis, cell proliferation, and growth, which might account for the positive effects of estrogen on the regeneration of motor neurons. Immunohistochemical staining revealed estrogen receptor-alpha and estrogen receptor-beta localized in the nucleus and cytoplasm of lumbar motor neurons, and in the regenerating neurites of the sciatic nerve. Expression of estrogen receptor-alpha and estrogen receptor-beta mRNA in lumbar spinal cord was shown by traditional RT-PCR. Using real-time quantitative RT-PCR, we demonstrated increased expression of estrogen receptors-alpha and -beta mRNA on the injured side of the lumbar spinal cord. Western blot analysis showed the accumulation of ERs in regenerating sciatic nerve, and revealed a 40% increase of activated ERK1/2 in estrogen treated mice, compared to placebo. Our findings indicate that: (i). axotomized motor neurons increase expression of estrogen receptors-alpha and -beta mRNA, (ii). estrogen mediates the expression of genes which accelerate the growth and maturation of axons, and (iii). estrogen receptors are transported from the perikaryon into regenerating neurites, and estrogen promotes regeneration locally through the non-genomic ERK-activated signaling pathway.
Collapse
Affiliation(s)
- Rustem R Islamov
- Department of Physiology, The Brody School of Medicine at East Carolina University, 600 Moye Blvd, Greenville, NC 27858, USA
| | | | | | | | | | | | | |
Collapse
|
90
|
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
|
91
|
Migliaccio A, Castoria G, Di Domenico M, de Falco A, Bilancio A, Lombardi M, Bottero D, Varricchio L, Nanayakkara M, Rotondi A, Auricchio F. Sex steroid hormones act as growth factors. J Steroid Biochem Mol Biol 2002; 83:31-5. [PMID: 12650699 DOI: 10.1016/s0960-0760(02)00264-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We observed that sex steroid hormones, like growth factors, stimulate the Src/Ras/erk pathway of cell lines derived from human mammary or prostate cancers. In addition, hormone-dependent pathway activation can be induced in Cos cells, upon transfection of classic steroid receptors. Cross-talks between sex steroid receptors regulate their association with Src and consequent pathway activation. Oestradiol treatment of MCF-7 cells triggers simultaneous association of ER with Src and p85, the regulatory subunit of phosphatidylinositol-3-kinase (PI3-kinase) and activation of Src- and PI3-K-dependent pathways. Activation of the latter pathway triggers cyclin D1 transcription, that is unaffected by Mek-1 activation. This suggests that simultaneous activation of different signalling effectors is required to target different cell cycle components. Thus, a novel reciprocal cross-talk between the two pathways appears to be mediated by the ER. In all tested cells, activation of the signalling pathways has a proliferative role. Transcriptionally inactive ER expressed in NIH 3T3 cells responds to hormone causing Src/Ras/Erk pathway activation and DNA synthesis. This suggests that in these cells genomic activity is required for later events of cell growth.
Collapse
Affiliation(s)
- A Migliaccio
- Dipartimento di Patologia generale-II Università di Napoli, Via L. De Crecchio, 7-I-80138 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Brownson DM, Azios NG, Fuqua BK, Dharmawardhane SF, Mabry TJ. Flavonoid effects relevant to cancer. J Nutr 2002; 132:3482S-3489S. [PMID: 12421874 DOI: 10.1093/jn/132.11.3482s] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Flavonoids, such as daidzein and genistein, present in dietary plants like soybean, have unique chemical properties with biological activity relevant to cancer. Many flavonoids and polyphenols, including resveratrol in red wine and epigallocatechin gallate in green tea, are known antioxidants. Some of these compounds have estrogenic (and antiestrogenic) activity and are commonly referred to as phytoestrogens. A yeast-based estrogen receptor (ER) reporter assay has been used to measure the ability of flavonoids to bind to ER and activate estrogen responsive genes. Recently, estrogenic compounds were also shown to trigger rapid, nongenomic effects. The molecular mechanisms, however, have not been completely detailed and little information exists regarding their relevance to cancer progression. As a preliminary step toward elucidating rapid phytoestrogen action on breast cancer cells, we investigated the effect of 17-beta estradiol (E2), genistein, daidzein and resveratrol on the activation status of signaling proteins that regulate cell survival and invasion, the cell properties underlying breast cancer progression. The effect of these estrogenic compounds on the activation, via phosphorylation, of Akt/protein kinase B (Akt) and focal adhesion kinase (FAK) were analyzed in ER-positive and -negative human breast cancer cell lines. E2, genistein and daidzein increased whereas resveratrol decreased both Akt and FAK phosphorylation in nonmetastatic ER-positive T47D cells. In metastatic ER-negative MDA-MB-231 cells, all estrogenic compounds tested increased Akt and FAK phosphorylation. The inhibitory action of resveratrol on cell survival and proliferation is ER dependent. Therefore, all estrogenic compounds tested, including resveratrol, may exert supplementary ER-independent nongenomic effects on cell survival and migration in breast cancer cells.
Collapse
Affiliation(s)
- Delia M Brownson
- Molecular Cell and Developmental Biology Section, The University of Texas at Austin, Austin, TX 78712, USA
| | | | | | | | | |
Collapse
|
93
|
Cardona-Gomez GP, Mendez P, Garcia-Segura LM. Synergistic interaction of estradiol and insulin-like growth factor-I in the activation of PI3K/Akt signaling in the adult rat hypothalamus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 107:80-8. [PMID: 12414126 DOI: 10.1016/s0169-328x(02)00449-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Estradiol and insulin-like growth factor-I (IGF-I) interact in the hypothalamus to regulate neuronal function, synaptic plasticity and neuroendocrine events. However, the molecular mechanisms involved in these interactions are still unknown. In the present study, the effect of estradiol on the signaling pathways of IGF-I receptor has been assessed in the hypothalamus of young adult ovariectomized rats, using specific antibodies for the phosphorylated forms of extracellular-signal regulated kinase (ERK) 1 and ERK2 and Akt/protein kinase B (Akt/PKB). Estradiol treatment resulted, between 6 and 24 h after systemic administration, in dose-dependent effects on the phosphorylation of ERK and Akt/PKB. Estradiol did not modify the level of ERK phosphorylation induced by intracerebroventricular administration of IGF-I. However, both hormones had a synergistic effect on the phosphorylation of Akt/PKB. These findings suggest that estrogen effects in the hypothalamus may be mediated in part by the activation of the signaling pathways of the IGF-I receptor.
Collapse
|
94
|
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
|
95
|
Wang L, Andersson S, Warner M, Gustafsson JA. Estrogen actions in the brain. SCIENCE'S STKE : SIGNAL TRANSDUCTION KNOWLEDGE ENVIRONMENT 2002; 2002:pe29. [PMID: 12084905 DOI: 10.1126/stke.2002.138.pe29] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Understanding of the mechanisms of estrogen action in the brain has always been poor. Neurons in several brain regions do not harbor estrogen receptor alpha (ERalpha) and yet are estrogen responsive. It was formerly thought that these responses represented indirect actions of estrogen. It is now evident that these neurons express ERbeta and that estrogen receptors have diverse actions in the central nervous system. By clear delineation of the cellular expression and function of the two estrogen receptors, it is likely that, in the future, selective ERalpha and ERbeta ligands will be developed and used for treatment of depression and behavioral disorders and may be useful in preventing degenerative diseases, such as Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Ling Wang
- Department of Medical Nutrition, Karolinska Institute, NOVUM, S-141 86 Huddinge, Sweden
| | | | | | | |
Collapse
|