首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
2.
3.
Neuroplasticity refers to the changes in the molecular and cellular processes of neural circuits that occur in response to environmental experiences. Clinical and experimental studies have increasingly shown that estrogens participate in the neuroplasticity involved in cognition, behavior, and memory. It is generally accepted that estrogens exert their effects through genomic actions that occur over a period of hours to days. However, emerging evidence indicates that estrogens also rapidly influence the neural circuitry through nongenomic actions. In this review, we provide an overview of the genomic and nongenomic actions of estrogens and discuss how these actions may cooperate in synaptic plasticity. We then summarize the role of epigenetic modifications, synaptic protein synthesis, and posttranslational modifications, and the splice variants of estrogen receptors in the complicated network of estrogens. The combination of genomic and nongenomic mechanisms endows estrogens with considerable diversity in modulating neural functions including synaptic plasticity.  相似文献   

4.
In addition to its well-known activational mechanism, the steroid hormone 17-beta-estradiol (E2) has been shown to rapidly activate various signal transduction pathways that could participate in estrogen-mediated regulation of synaptic plasticity. Although the mechanisms underlying these effects are not clearly understood, it has been repeatedly suggested that they involve a plasma membrane receptor which has direct links to several intracellular signaling cascades. To further address the question of whether E2 acts directly at the synapse and through membrane-bound receptors, we studied the effects of E2 and of ligands of estrogen receptors on various signaling pathways in cortical synaptoneurosomes. Our results demonstrate that E2 elicits N-methyl-D-aspartate receptor phosphorylation and activates the extracellular signal-regulated kinase and the phosphatidylinositol 3-kinase/Akt signal transduction pathways in this cortical membrane preparation. Furthermore, we provide evidence for the presence of a membrane-bound estrogen receptor responsible for these effects in cortical synaptoneurosomes. Our study demonstrates that E2 directly acts at cortical synapses, and that synaptoneurosomes provide a useful system to investigate the mechanisms by which E2 regulates synaptic transmission and plasticity.  相似文献   

5.
Extra-nuclear signaling of estrogen receptors   总被引:1,自引:0,他引:1  
Fu XD  Simoncini T 《IUBMB life》2008,60(8):502-510
  相似文献   

6.
Estrogen is an important hormone signal that regulates multiple tissues and functions in the body. This review focuses on the neurotrophic and neuroprotective actions of estrogen in the brain, with particular emphasis on estrogen actions in the hippocampus, cerebral cortex and striatum. Sex differences in the risk, onset and severity of neurodegenerative disease such as Alzheimer's disease, Parkinson's disease and stroke are well known, and the potential role of estrogen as a neuroprotective factor is discussed in this context. The review assimilates a complex literature that spans research in humans, non-human primates and rodent animal models and attempts to contrast and compare the findings across species where possible. Current controversies regarding the Women's Health Initiative (WHI) study, its ramifications, concerns and the new studies needed to address these concerns are also addressed. Signaling mechanisms underlying estrogen-induced neuroprotection and synaptic plasticity are reviewed, including the important concepts of genomic versus nongenomic mechanisms, types of estrogen receptor involved and their subcellular targeting, and implicated downstream signaling pathways and mediators. Finally, a multicellular mode of estrogen action in the regulation of neuronal survival and neurotrophism is discussed, as are potential future directions for the field.  相似文献   

7.
8.
9.
In the brain, membrane associated nongenomic steroid receptors can induce fast-acting responses to ion conductance and second messenger systems of neurons. Emerging data suggest that membrane associated glucocorticoid and mineralocorticoid receptors may directly regulate synaptic excitability during times of stress when adrenal hormones are elevated. As the key neuron signaling interface, the synapse is involved in learning and memory, including traumatic memories during times of stress. The lateral amygdala is a key site for synaptic plasticity underlying conditioned fear, which can both trigger and be coincident with the stress response. A large body of electrophysiological data shows rapid regulation of neuronal excitability by steroid hormone receptors. Despite the importance of these receptors, to date, only the glucocorticoid receptor has been anatomically localized to the membrane. We investigated the subcellular sites of mineralocorticoid receptors in the lateral amygdala of the Sprague-Dawley rat. Immunoblot analysis revealed the presence of mineralocorticoid receptors in the amygdala. Using electron microscopy, we found mineralocorticoid receptors expressed at both nuclear including: glutamatergic and GABAergic neurons and extra nuclear sites including: presynaptic terminals, neuronal dendrites, and dendritic spines. Importantly we also observed mineralocorticoid receptors at postsynaptic membrane densities of excitatory synapses. These data provide direct anatomical evidence supporting the concept that, at some synapses, synaptic transmission is regulated by mineralocorticoid receptors. Thus part of the stress signaling response in the brain is a direct modulation of the synapse itself by adrenal steroids.  相似文献   

10.
Cell morphology is controlled by a complex and redundant array of intracellular signaling pathways devoted to the regulation of the actin cytoskeleton and of its relationship with the cell membrane and the extracellular matrix. Sex steroids are effective regulators of cell morphology and tissue organization, and recent evidence indicates that this is obtained through the regulation of the cytoskeleton. Intriguingly, many of these regulatory actions related to cell morphology are achieved through rapid, non-classical signaling of sex steroid receptors to kinase cascades, independently from nuclear alteration of gene expression or protein synthesis. The identification of the mechanistic basis for these rapid actions on cell cytoskeleton has special relevance for the characterization of the effects of sex steroids in physiological conditions, such as their role in the control of brain cell remodeling. Brain cell morphology is controlled by estrogens that regulate the development of neuron/neuron interconnections and dendritic spine density. This is thought to be critical for gender-specific differences in brain function and dysfunction. The recent advancements in the characterization of the molecular basis of the extra-nuclear signaling of estrogen helps to understand the role of estrogen in the brain, and may in the future turn out to be of relevance for clinical purposes. This review highlights the regulatory effects on the cytoskeleton and cell morphology of estrogens as well as the recent advances in the characterization of these mechanisms, providing insights and working hypotheses on possible clinical applications for the modulation of these pathways in the central nervous system.  相似文献   

11.
12.
Herrera JL  Fernandez C  Diaz M  Cury D  Marin R 《Steroids》2011,76(9):840-844
There is a wealth of information indicating that estradiol exerts rapid actions involved in neuroprotection and cognitive-enhancing effects. Some of these effects appear to delay onset, or even ameliorate, the neuropathology of Alzheimer's disease (AD), although some controversy exists about the beneficial brain effects of estrogen therapies. Therefore, it is crucial to better understand the mechanisms developed by 17β-estradiol to signal in the brain. At the neuronal membrane, the hormone can rapidly interact with estrogen receptors (mERs) or activate other receptors, such as G protein-coupled and ionotropic receptors. And the list of membrane signalling molecules modulated by estradiol in neurons is increasing. VDAC is a voltage-dependent anion channel, known as a mitochondrial porin which is also found at the neuronal membrane, where it appears to be involved in redox regulation, extrinsic apoptosis and amyloid beta neurotoxicity. Moreover, VDAC is present in neuronal lipid rafts, where it is associated with estrogen receptor α-like (mER), forming part of a macromolecular complex together with caveolin-1 and other signalling proteins related to neuronal preservation. Interestingly, we have recently found that 17β-estradiol rapidly promotes VDAC phosphorylation through the activation of protein kinase A (PKA) and Src-kinase, which may be relevant to maintain this channel inactivated. On the contrary, tamoxifen, a selective estrogen receptor modulator (SERM), provokes the dephosphorylation of VDAC, and eventually its opening, by activating a cascade of phosphatases, including protein phosphatase 2 (PP2A). This review will focus on the relevance of these novel findings in the alternative estrogen mechanisms to achieve neuroprotection related to AD.  相似文献   

13.
The potential neuroprotective role of sex hormones in chronic neurodegenerative disorders and acute brain ischemia following cardiac arrest and stroke is of a great therapeutic interest. Long-term pretreatment with estradiol and other estrogens affords robust neuroprotection in male and female rodents subjected to focal and global ischemia. However, the receptors (e.g., cell surface or nuclear), intracellular signaling pathways and networks of estrogen-regulated genes that intervene in neuronal apoptosis are as yet unclear. We have shown that estradiol administered at physiological levels for two weeks before ischemia rescues neurons destined to die in the hippocampal CA1 and ameliorates ischemia-induced cognitive deficits in ovariectomized female rats. This regimen of estradiol treatment involves classical intracellular estrogen receptors, transactivation of IGF-1 receptors and stimulation of the ERK/MAPK signaling pathway, which in turn maintains CREB activity in the ischemic CA1. We also find that a single, acute injection of estradiol administrated into the brain ventricle immediately after an ischemic event reduces both neuronal death and cognitive deficits. Because these findings suggest that hormones could be used to treat patients when given after brain ischemia, it is critical to determine whether the same or different pathways mediate this form of neuroprotection. We find that an agonist of the membrane estrogen receptor GPR30 mimics short latency estradiol facilitation of synaptic transmission in the hippocampus. Therefore, we are testing the hypothesis that GPR30 may act together with intracellular estrogen receptors to activate cell signaling pathways to promote neuron survival after global ischemia.  相似文献   

14.
15.
A large body of evidence supports a role for the NO-cGMP-protein kinase G pathway in the regulation of synaptic transmission and plasticity, brain development and neuroprotection. Circumstancial evidence implicates natriuretic peptide-stimulated cGMP formation in the same CNS functions. In addition to neurons, both cGMP-mediated pathways are functional in glial cells and an increasing number of reports indicate that they may control important aspects of glial cell physiology relevant to neuronal function. In this article we briefly review the regulation of cGMP formation in glial cells and summarize recent evidence indicating that cGMP-mediated pathways can play important roles in astroglial and microglial function in normal and diseased brain. Special issue article in honor of Dr. Anna Maria Giuffrida-Stella.  相似文献   

16.
17.
Membrane initiated estrogen signaling in breast cancer   总被引:1,自引:0,他引:1  
Recent research has focused on effects of the estrogen receptor acting at the level of the cell membrane in breast cancer. In this review we describe 17beta-estradiol (E2)-initiated membrane signaling pathways involving the activation of several kinases that contribute to the regulation of cell proliferation and prevention of apoptosis. Although classical concepts had assigned priority to the nuclear actions of estrogen receptor, recent studies document the additional importance of estrogen receptor residing in or near the plasma membrane. A small fraction of estrogen receptor is associated with the cell membrane and mediates the rapid effects of E2. Unlike classical growth factor receptors, such as insulin-like growth factor 1 receptor (IGF1R) and epidermal growth factor receptor (EGFR), estrogen receptor has no transmembrane and kinase domains and is known to initiate E2 rapid signals by forming a protein complex with many signaling molecules. The formation of the protein complex is a critical step, leading to the activation of the MAPK1/3 (also known as MAP kinase) and AKT1 (also known as Akt) pathways. A full understanding of the mechanisms underlying these relationships, with the ultimate aim of abrogating specific steps, should lead to more-targeted strategies for treatment of hormone dependent-breast cancer.  相似文献   

18.
Integrin class adhesion proteins are concentrated at adult brain synapses. Whether synaptic integrins engage kinase signaling cascades has not been determined, but is a question of importance to ideas about integrin involvement in functional synaptic plasticity. Accordingly, synaptoneurosomes from adult rat brain were used to test if matrix ligands activate integrin-associated tyrosine kinases, and if integrin signaling targets include NMDA-class glutamate neurotransmitter receptors. The integrin ligand peptide Gly-Arg-Gly-Asp-Ser-Pro (GRGDSP) induced rapid (within 5 min) and robust increases in tyrosine phosphorylation of focal adhesion kinase, proline-rich tyrosine kinase 2 and Src family kinases. Increases were similarly induced by the native ligand fibronectin, blocked with neutralizing antibodies to beta1 integrin, and not obtained with control peptides, indicating that kinase activation was integrin-mediated. Both GRGDSP and fibronectin caused rapid Src kinase-dependent increases in tyrosine phosphorylation of NMDA receptor subunits NR2A and NR2B in synaptoneurosomes and acute hippocampal slices. Tests of the physiological significance of the latter result showed that ligand treatment caused a rapid and beta1 integrin-dependent increase in NMDA receptor-mediated synaptic responses. These results provide the first evidence that, in adult brain, synaptic integrins activate local kinase cascades with potent effects on the operation of nearby neurotransmitter receptors implicated in synaptic plasticity.  相似文献   

19.
Estradiol (E(2)) exerts not only genotropic but also nongenomic actions through nuclear estrogen receptors (ER). Here, we provide a novel paradigm for nongenomic E(2) signaling independent of nuclear ER. E(2) induces a rapid rise in the intracellular free Ca(2+) concentration ([Ca(2+)](i)) through membrane estrogen receptors in murine RAW 264.7 macrophages. This E(2)-induced Ca(2+) signaling is not prevented by different ER blockers and cannot directly activate stably transfected c-fos promoter or the mitogen-activated protein kinases p38, ERK1/2, and SAPK/JNK, or NO production. However, the E(2)-induced rise in [Ca(2+)](i) specifically down-regulates the serum-stimulated activation of c-fos promoter and ERK1/2, and conversely, it specifically up-regulates lipopolysaccharide-stimulated activation of c-fos promoter, p38, and NO production. The E(2)-changed activation of c-fos promoter can be prevented by an intracellular Ca(2+) chelator. Our data indicate that E(2)-induced nongenomic Ca(2+) signaling through membrane ER is able to specifically modulate genotropic signaling pathways with impact on macrophage activation.  相似文献   

20.
Giretti MS  Simoncini T 《Steroids》2008,73(9-10):895-900
Cell movement is required in relevant physiological processes such as embryonic development, tissue and organ differentiation, inflammation, immune response and wound healing, along with pathological phenomena, such as cancer metastatic spread. Cell motility is tightly controlled by a complex and often redundant array of intracellular signaling pathways largely devoted to the dynamic regulation of the actin cytoskeletal network and of its relationship with the cell membrane and the extracellular matrix. Sex steroids, particularly estrogen and progesterone, are effective regulators of cell migration and tissue organization, and recent evidence indicates that this is in part obtained through the regulation of the cytoskeleton. Intriguingly, many of these regulatory actions related to cell movement are achieved through rapid, non-classical signaling of sex steroid receptors to kinase cascades, independently from nuclear alteration of gene expression or protein synthesis. The identification of the mechanistic basis for these rapid actions on cell cytoskeleton and cell movement has special relevance for the characterization of the effects of sex steroids in physiological conditions, such their role in the control of inflammation, brain or vascular cell remodelling, angiogenesis or wound healing, as well as in the context of pathological conditions such as steroid-sensitive cancer cell invasion and metastasis. This review highlights the physiological and clinical conditions where the regulatory effects on the cytoskeleton and cell movement of sex steroids might have a special importance, as well as the recent advances in the characterization of the mechanisms, providing insights and working hypotheses on possible clinical applications for the modulation of these pathways.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号