1
|
Taxier LR, Pillerová M, Branyan TE, Sohrabji F, Frick KM. Astrocytic glutamate transport is essential for the memory-enhancing effects of 17β-estradiol in ovariectomized mice. Horm Behav 2024; 165:105618. [PMID: 39180889 PMCID: PMC11498968 DOI: 10.1016/j.yhbeh.2024.105618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/25/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
Infusion of 17β-estradiol (E2) into the dorsal hippocampus (DH) of ovariectomized (OVX) mice enhances memory consolidation, an effect that depends on rapid phosphorylation of extracellular signal-regulated kinase (ERK) and Akt. Astrocytic glutamate transporter 1 (GLT-1) modulates neurotransmission via glutamate uptake from the synaptic cleft. However, little is known about the contribution of DH astrocytes, and astrocytic glutamate transport, to the memory-enhancing effects of E2. This study was designed to test whether DH astrocytes contribute to estrogenic modulation of memory consolidation by determining the extent to which DH GLT-1 is necessary for E2 to enhance memory in object recognition and object placement tasks and trigger rapid phosphorylation events in DH astrocytes. OVX female mice were bilaterally cannulated into the DH or the DH and dorsal third ventricle (ICV). Post-training DH infusion of the GLT-1 inhibitor dihydrokainic acid (DHK) dose-dependently impaired memory consolidation in both tasks. Moreover, the memory-enhancing effects of ICV-infused E2 in each task were blocked by DH DHK infusion. E2 increased p42 ERK and Akt phosphorylation in DH astrocytes, and these effects were blocked by DHK. Results suggest the necessity of DH GLT-1 activity for object and spatial memory consolidation, and for E2 to enhance consolidation of these memories and to rapidly activate cell signaling in DH astrocytes. Findings indicate that astrocytic function in the DH of OVX females is necessary for memory formation and is regulated by E2, and suggest an essential role for DH astrocytic GLT-1 activity in the memory-enhancing effects of E2.
Collapse
Affiliation(s)
- Lisa R Taxier
- University of Wisconsin-Milwaukee, Department of Psychology, Milwaukee, WI, USA.
| | - Miriam Pillerová
- University of Wisconsin-Milwaukee, Department of Psychology, Milwaukee, WI, USA; Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic.
| | - Taylor E Branyan
- Texas A&M Institute for Neuroscience and TAMU College of Medicine, Bryan, TX, USA.
| | - Farida Sohrabji
- Texas A&M Institute for Neuroscience and TAMU College of Medicine, Bryan, TX, USA.
| | - Karyn M Frick
- University of Wisconsin-Milwaukee, Department of Psychology, Milwaukee, WI, USA.
| |
Collapse
|
2
|
Cui XW, Ren JY, Gu YH, Li QF, Wang ZC. NF1, Neurofibromin and Gene Therapy: Prospects of Next-Generation Therapy. Curr Gene Ther 2020; 20:100-108. [PMID: 32767931 DOI: 10.2174/1566523220666200806111451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 12/28/2022]
Abstract
Neurofibromatosis type 1 [NF1] is an autosomal dominant genetic disorder affecting multiple organs. NF1 is well known for its various clinical manifestations, including café-au-late macules, Lisch nodules, bone deformity and neurofibromas. However, there is no effective therapy for NF1. Current therapies are aimed at alleviating NF1 clinical symptoms but not curing the disease. By altering pathogenic genes, gene therapy regulates cell activities at the nucleotide level. In this review, we described the structure and functions of neurofibromin domains, including GAP-related domain [GRD], cysteine-serine rich domain [CSRD], leucine-rich domain [LRD] and C-terminal domain [CTD], which respectively alter downstream pathways. By transfecting isolated sequences of these domains, researchers can partially restore normal cell functions in neurofibroma cell lines. Furthermore, recombinant transgene sequences may be designed to encode truncated proteins, which is functional and easy to be packaged into viral vectors. In addition, the treatment effect of gene therapy is also determined by various factors such as the vectors selection, transgene packaging strategies and drug administration. We summarized multiple NF1 gene therapy strategies and discussed their feasibility from multiple angles. Different protein domains alter the function and downstream pathways of neurofibromin.
Collapse
Affiliation(s)
- Xi-Wei Cui
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jie-Yi Ren
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi-Hui Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhi-Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
3
|
Kang M, Lee YS. The impact of RASopathy-associated mutations on CNS development in mice and humans. Mol Brain 2019; 12:96. [PMID: 31752929 PMCID: PMC6873535 DOI: 10.1186/s13041-019-0517-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/28/2019] [Indexed: 01/04/2023] Open
Abstract
The RAS signaling pathway is involved in the regulation of developmental processes, including cell growth, proliferation, and differentiation, in the central nervous system (CNS). Germline mutations in the RAS signaling pathway genes are associated with a group of neurodevelopmental disorders, collectively called RASopathy, which includes neurofibromatosis type 1, Noonan syndrome, cardio-facio-cutaneous syndrome, and Costello syndrome. Most mutations associated with RASopathies increase the activity of the RAS-ERK signaling pathway, and therefore, most individuals with RASopathies share common phenotypes, such as a short stature, heart defects, facial abnormalities, and cognitive impairments, which are often accompanied by abnormal CNS development. Recent studies using mouse models of RASopathies demonstrated that particular mutations associated with each disorder disrupt CNS development in a mutation-specific manner. Here, we reviewed the recent literatures that investigated the developmental role of RASopathy-associated mutations using mutant mice, which provided insights into the specific contribution of RAS-ERK signaling molecules to CNS development and the subsequent impact on cognitive function in adult mice.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.
| |
Collapse
|
4
|
de Almeida AA, Gomes da Silva S, Lopim GM, Vannucci Campos D, Fernandes J, Cabral FR, Arida RM. Physical exercise alters the activation of downstream proteins related to BDNF-TrkB signaling in male Wistar rats with epilepsy. J Neurosci Res 2017; 96:911-920. [DOI: 10.1002/jnr.24196] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Alexandre Aparecido de Almeida
- Departamento de Fisiologia; Universidade Federal de São Paulo; São Paulo Brazil
- Instituto Federal Goiano (IF Goiano), Campus Ceres; Ceres Brazil
| | - Sérgio Gomes da Silva
- Hospital Israelita Albert Einstein; São Paulo Brazil
- Universidade de Mogi das Cruzes; Mogi das Cruzes Brazil
| | | | | | - Jansen Fernandes
- Departamento de Fisiologia; Universidade Federal de São Paulo; São Paulo Brazil
| | - Francisco Romero Cabral
- Hospital Israelita Albert Einstein; São Paulo Brazil
- Faculdade de Ciências Médicas da Santa Casa de São Paulo; São Paulo Brazil
| | - Ricardo Mario Arida
- Departamento de Fisiologia; Universidade Federal de São Paulo; São Paulo Brazil
| |
Collapse
|
5
|
Inhibition of miR-181a protects female mice from transient focal cerebral ischemia by targeting astrocyte estrogen receptor-α. Mol Cell Neurosci 2017; 82:118-125. [PMID: 28522364 DOI: 10.1016/j.mcn.2017.05.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/03/2017] [Accepted: 05/08/2017] [Indexed: 02/03/2023] Open
Abstract
Whether the effect of miR-181a is sexually dimorphic in stroke is unknown. Prior work showed protection of male mice with miR-181a inhibition. Estrogen receptor-α (ERα) is an identified target of miR181 in endometrium. Therefore we investigated the separate and joint effects of miR-181a inhibition and 17β-estradiol (E2) replacement after ovariectomy. Adult female mice were ovariectomized and implanted with an E2- or vehicle-containing capsule for 14d prior to 1h middle cerebral artery occlusion (MCAO). Each group received either miR-181a antagomir or mismatch control by intracerebroventricular injection 24h before MCAO. After MCAO neurologic deficit and infarct volume were assessed. Primary male and female astrocyte cultures were subjected to glucose deprivation with miR-181a inhibitor or transfection control, and E2 or vehicle control, with/without ESRα knockdown with small interfering RNA. Cell death was assessed by propidium iodide staining, and lactate dehydrogenase assay. A miR-181a/ERα target site blocker (TSB), with/without miR-181a mimic, was used to confirm targeting of ERα by miR-181a in astrocytes. Individually, miR-181a inhibition or E2 decreased infarct volume and improved neurologic score in female mice, and protected male and female astrocyte cultures. Combined miR-181a inhibition plus E2 afforded greater protection of female mice and female astrocyte cultures, but not in male astrocyte cultures. MiR-181a inhibition only increased ERα levels in vivo and in female cultures, while ERα knockdown with siRNA increased cell death in both sexes. Treatment with ERα TSB was strongly protective in both sexes. In conclusion, the results of the present study suggest miR-181a inhibition enhances E2-mediated stroke protection in females in part by augmenting ERα production, a mechanism detected in female mice and female astrocytes. Sex differences were observed with combined miR-181a inhibition/E2 treatment, and miR-181a targeting of ERα.
Collapse
|
6
|
Yi H, Bao X, Tang X, Fan X, Xu H. Estrogen modulation of calretinin and BDNF expression in midbrain dopaminergic neurons of ovariectomised mice. J Chem Neuroanat 2016; 77:60-67. [PMID: 27211874 DOI: 10.1016/j.jchemneu.2016.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 05/09/2016] [Accepted: 05/18/2016] [Indexed: 11/19/2022]
Abstract
Estrogen attenuates the loss of dopamine neurons from the substantia nigra in animal models of Parkinson's disease (PD) and excitatory amino-acid induced neurotoxicity by interactions with brain-derived neurotrophic factor (BDNF), and calretinin (CR) containing dopaminergic (DA) neurons. To examine this interaction more closely, we treated the ovariectomised (OVX) mice with estrodial for 10days, and compared these mice to those OVX mice injected with the vehicle or control mice. Estrogen treatment in OVX mice had significantly more tyrosine hydroxylase (TH) positive neurons in the substantia nigra pars compacta (SNpc). Dopamine transporter (DAT) mRNA and BDNF mRNA levels in the midbrain were also significantly increased by estrogen treatment (P<0.05). OVX markedly decreased the number of TH/CR double stained cells in the SNpc (P<0.05), a trend which could be reversed by estrogen treatment. However, the number of GFAP positive cells in the substantia nigra did not show significant changes (P >0.05) after vehicle or estrodial treatment. Furthermore, we found that estrogen treatment abrogated the OVX-induced decrease in the phosphorylated AKT (p-AKT), but not p-ERK. We hypothesize that short-term treatment with estrogen confers neuroprotection to DA neurons by increasing CR in the DA neurons and BDNF in the midbrain, which possibly related to activation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Hongliang Yi
- Department of Physiology, Third Military Medical University, Chongqing, 400038, PR China; Chongqing City Family Planning Institute, Chongqing, 400020, PR China
| | - Xiaohang Bao
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Xiaotong Tang
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, PR China
| | - Xiaotang Fan
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital,Third Military Medical University, Chongqing, 400038, PR China.
| |
Collapse
|
7
|
Gonsalvez D, Ferner AH, Peckham H, Murray SS, Xiao J. The roles of extracellular related-kinases 1 and 2 signaling in CNS myelination. Neuropharmacology 2015; 110:586-593. [PMID: 25959068 DOI: 10.1016/j.neuropharm.2015.04.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/16/2015] [Accepted: 04/27/2015] [Indexed: 01/09/2023]
Abstract
Substantial progress has been made in identifying the intracellular signaling pathways that regulate central nervous system myelination. Recently, the mitogen activated protein kinase pathway, in particular the extracellular signal-related kinase 1 (Erk1) and Erk2, have been identified as critically important in mediating the effects of several growth factors that regulate oligodendroglial development and myelination. Here we will review the recent studies that identify the key role that Erk1/2 signaling plays in regulating oligodendroglial development, myelination and remyelination, discuss the potential mechanisms that Erk1/2 may utilize to influence myelination, and highlight some questions for further research. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- David Gonsalvez
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Anita H Ferner
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Haley Peckham
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Simon S Murray
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia; The Florey Institute of Neuroscience and Mental Health Research, The University of Melbourne, Victoria 3010, Australia
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia; The Florey Institute of Neuroscience and Mental Health Research, The University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
8
|
Benmansour S, Privratsky AA, Adeniji OS, Frazer A. Signaling mechanisms involved in the acute effects of estradiol on 5-HT clearance. Int J Neuropsychopharmacol 2014; 17:765-77. [PMID: 24423185 PMCID: PMC3969768 DOI: 10.1017/s146114571300165x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Estradiol was found previously to have an antidepressant-like effect and to block the ability of selective serotonin reuptake inhibitors (SSRIs) to have an antidepressant-like effect. The antidepressant-like effect of estradiol was due to estrogen receptor β (ERβ) and/or GPR30 activation, whereas estradiol's blockade of the effect of an SSRI was mediated by ERα. This study focuses on investigating signaling pathways as well as interacting receptors associated with these two effects of estradiol. In vivo chronoamperometry was used to measure serotonin transporter (SERT) function. The effect of local application of estradiol or selective agonists for ERα (PPT) or ERβ (DPN) into the CA3 region of the hippocampus of ovariectomized (OVX) rats on 5-hydroxytryptamine (5-HT) clearance as well as on the ability of fluvoxamine to slow 5-HT clearance was examined after selective blockade of signaling pathways or that of interacting receptors. Estradiol- or DPN-induced slowing of 5-HT clearance mediated by ERβ was blocked after inhibition of MAPK/ERK1/2 but not of PI3K/Akt signaling pathways. This effect also involved interactions with TrkB, and IGF-1 receptors. Estradiol's or PPT's inhibition of the fluvoxamine-induced slowing of 5-HT clearance mediated by ERα, was blocked after inhibition of either MAPK/ERK1/2 or PI3K/Akt signaling pathways. This effect involved interactions with the IGF-1 receptor and with the metabotropic glutamate receptor 1, but not with TrkB. This study illustrates some of the signaling pathways required for the effects of estradiol on SERT function, and particularly shows that ER subtypes elicit different as well as common signaling pathways for their actions.
Collapse
Affiliation(s)
- Saloua Benmansour
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Anthony A. Privratsky
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Opeyemi S. Adeniji
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Alan Frazer
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
- South Texas Veterans Health Care System, San Antonio, Texas 78284, USA
| |
Collapse
|
9
|
Clark S, Rainville J, Zhao X, Katzenellenbogen BS, Pfaff D, Vasudevan N. Estrogen receptor-mediated transcription involves the activation of multiple kinase pathways in neuroblastoma cells. J Steroid Biochem Mol Biol 2014; 139:45-53. [PMID: 24121066 DOI: 10.1016/j.jsbmb.2013.09.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 09/15/2013] [Accepted: 09/20/2013] [Indexed: 11/22/2022]
Abstract
While many physiological effects of estrogens (E) are due to regulation of gene transcription by liganded estrogen receptors (ERs), several effects are also mediated, at least in part, by rapid non-genomic actions of E. Though the relative importance of rapid versus genomic effects in the central nervous system is controversial, we showed previously that membrane-limited effects of E, initiated by an estradiol bovine serum albumin conjugate (E2-BSA), could potentiate transcriptional effects of 17β-estradiol from an estrogen response element (ERE)-reporter in neuroblastoma cells. Here, using specific inhibitors and activators in a pharmacological approach, we show that activation of phosphatidylinositol-3-phosphate kinase (PI3K) and mitogen activated protein kinase (MAPK) pathways, dependent on a Gαq coupled receptor signaling are important in this transcriptional potentiation. We further demonstrate, using ERα phospho-deficient mutants, that E2-BSA mediated phosphorylation of ERα is one mechanism to potentiate transcription from an ERE reporter construct. This study provides a possible mechanism by which signaling from the membrane is coupled to transcription in the nucleus, providing an integrated view of hormone signaling in the brain.
Collapse
Affiliation(s)
- Sara Clark
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, United States
| | | | | | | | | | | |
Collapse
|
10
|
Cheng P, Alberts I, Li X. The role of ERK1/2 in the regulation of proliferation and differentiation of astrocytes in developing brain. Int J Dev Neurosci 2013; 31:783-9. [DOI: 10.1016/j.ijdevneu.2013.09.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 09/19/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022] Open
Affiliation(s)
- Peipei Cheng
- Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ian Alberts
- Department of Natural Sciences, LaGuardia CCCity University of New YorkNY11101USA
| | - Xiaohong Li
- Department of NeurochemistryNY State Institute for Basic Research in Developmental DisabilitiesNew YorkNY10314USA
| |
Collapse
|
11
|
Involvement of IGF-I receptor and estrogen receptor pathways in the protective effects of ginsenoside Rg1 against Aβ25–35-induced toxicity in PC12 cells. Neurochem Int 2013; 62:1065-71. [DOI: 10.1016/j.neuint.2013.03.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 03/20/2013] [Accepted: 03/29/2013] [Indexed: 12/20/2022]
|
12
|
Sinchak K, Wagner EJ. Estradiol signaling in the regulation of reproduction and energy balance. Front Neuroendocrinol 2012; 33:342-63. [PMID: 22981653 PMCID: PMC3496056 DOI: 10.1016/j.yfrne.2012.08.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/18/2012] [Accepted: 08/22/2012] [Indexed: 12/14/2022]
Abstract
Our knowledge of membrane estrogenic signaling mechanisms and their interactions that regulate physiology and behavior has grown rapidly over the past three decades. The discovery of novel membrane estrogen receptors and their signaling mechanisms has started to reveal the complex timing and interactions of these various signaling mechanisms with classical genomic steroid actions within the nervous system to regulate physiology and behavior. The activation of the various estrogenic signaling mechanisms is site specific and differs across the estrous cycle acting through both classical genomic mechanisms and rapid membrane-initiated signaling to coordinate reproductive behavior and physiology. This review focuses on our current understanding of estrogenic signaling mechanisms to promote: (1) sexual receptivity within the arcuate nucleus of the hypothalamus, (2) estrogen positive feedback that stimulates de novo neuroprogesterone synthesis to trigger the luteinizing hormone surge important for ovulation and estrous cyclicity, and (3) alterations in energy balance.
Collapse
Affiliation(s)
- Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, 1250 Bellflower Blvd., Long Beach, CA 90840-9502, United States.
| | | |
Collapse
|
13
|
Kuo J, Micevych P. Neurosteroids, trigger of the LH surge. J Steroid Biochem Mol Biol 2012; 131:57-65. [PMID: 22326732 PMCID: PMC3474707 DOI: 10.1016/j.jsbmb.2012.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 01/19/2012] [Accepted: 01/22/2012] [Indexed: 12/28/2022]
Abstract
Recent experiments from our laboratory are consistent with the idea that hypothalamic astrocytes are critical components of the central nervous system (CNS) mediated estrogen positive feedback mechanism. The "astrocrine hypothesis" maintains that ovarian estradiol rapidly increases free cytoplasmic calcium concentrations ([Ca(2+)](i)) that facilitate progesterone synthesis in astrocytes. This hypothalamic neuroprogesterone along with the elevated estrogen from the ovaries allows for the surge release of gonadotropin-releasing hormone (GnRH) that triggers the pituitary luteinizing hormone (LH) surge. A narrow range of estradiol stimulated progesterone production supports an "off-on-off" mechanism regulating the transition from estrogen negative feedback to estrogen positive feedback, and back again. The rapidity of the [Ca(2+)](i) response and progesterone synthesis support a non-genomic, membrane-initiated signaling mechanism. In hypothalamic astrocytes, membrane-associated estrogen receptors (mERs) signal through transactivation of the metabotropic glutamate receptor type 1a (mGluR1a), implying that astrocytic function is influenced by surrounding glutamatergic nerve terminals. Although other putative mERs, such as mERβ, STX-activated mER-Gα(q), and G protein-coupled receptor 30 (GPR30), are present and participate in membrane-mediated signaling, their influence in reproduction is still obscure since female reproduction be it estrogen positive feedback or lordosis behavior requires mERα. The astrocrine hypothesis is also consistent with the well-known sexual dimorphism of estrogen positive feedback. In rodents, only post-pubertal females exhibit this positive feedback. Hypothalamic astrocytes cultured from females, but not males, responded to estradiol by increasing progesterone synthesis. Estrogen autoregulates its own signaling by regulating levels of mERα in the plasma membrane of female astrocytes. In male astrocytes, the estradiol-induced increase in mERα was attenuated, suggesting that membrane-initiated estradiol signaling (MIES) would also be blunted. Indeed, estradiol induced [Ca(2+)](i) release in male astrocytes, but not to levels required to stimulate progesterone synthesis. Investigation of this sexual differentiation was performed using hypothalamic astrocytes from post-pubertal four core genotype (FCG) mice. In this model, genetic sex is uncoupled from gonadal sex. We demonstrated that animals that developed testes (XYM and XXM) lacked estrogen positive feedback, strongly suggesting that the sexual differentiation of progesterone synthesis is driven by the sex steroid environment during early development. This article is part of a Special Issue entitled 'Neurosteroids'.
Collapse
Affiliation(s)
- John Kuo
- Department of Neurobiology, Laboratory of Neuroendocrinology of the Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - Paul Micevych
- Department of Neurobiology, Laboratory of Neuroendocrinology of the Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
- Corresponding author at: Department of Neurobiology, David Geffen School of Medicine at UCLA, 10833 LeConte Avenue, 73-078 CHS, Los Angeles, CA 90095-1763, United States. Tel.: +1 310 206 8265; fax: +1 310 825 2224. (P. Micevych)
| |
Collapse
|
14
|
Barreto GE, Gonzalez J, Capani F, Morales L. Neuroprotective agents in brain injury: a partial failure? Int J Neurosci 2012; 122:223-6. [PMID: 22176297 DOI: 10.3109/00207454.2011.648292] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Brain injury leads to inflammation, stress, and cell death. Neurons are more susceptible to injury than astrocytes, as they have limited antioxidant capacity, and rely heavily on their metabolic coupling with astrocytes to combat oxidative stress. Both normally and after brain injury, astrocytes support neurons by providing antioxidant protection, substrates for neuronal metabolism, and glutamate clearance. Although astrocytes are generally more resilient than neurons after injury, severe damage also results in astrocyte dysfunction, leading to increased neuronal death. This mini review provides a very insightful and brief overview on a few examples of promising neuroprotective compounds targeting astrocyte function, with specific attention on how these treatments alter astrocyte response or viability, and how this may be critical for neuronal survival following brain injury.
Collapse
Affiliation(s)
- George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia.
| | | | | | | |
Collapse
|
15
|
Micevych P, Sinchak K. The Neurosteroid Progesterone Underlies Estrogen Positive Feedback of the LH Surge. Front Endocrinol (Lausanne) 2011; 2:90. [PMID: 22654832 PMCID: PMC3356049 DOI: 10.3389/fendo.2011.00090] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 11/16/2011] [Indexed: 01/25/2023] Open
Abstract
Our understanding the steroid regulation of neural function has rapidly evolved in the past decades. Not long ago the prevailing thoughts were that peripheral steroid hormones carried information to the brain which passively responded to these steroids. These steroid actions were slow, taking hours to days to be realized because they regulated gene expression. Over the past three decades, discoveries of new steroid receptors, rapid membrane-initiated signaling mechanisms, and de novo neurosteroidogenesis have shed new light on the complexity of steroids actions within the nervous system. Sexual differentiation of the brain during development occurs predominately through timed steroid-mediated expression of proteins and long term epigenetic modifications. In contrast across the estrous cycle, estradiol release from developing ovarian follicles initially increases slowly and then at proestrus increases rapidly. This pattern of estradiol release acts through both classical genomic mechanisms and rapid membrane-initiated signaling in the brain to coordinate reproductive behavior and physiology. This review focuses on recently discovered estrogen receptor-α membrane signaling mechanisms that estradiol utilizes during estrogen positive feedback to stimulate de novo progesterone synthesis within the hypothalamus to trigger the luteinizing hormone (LH) surge important for ovulation and estrous cyclicity. The activation of these signaling pathways appears to be coordinated by the rising and waning of estradiol throughout the estrous cycle and integral to the negative and positive feedback mechanisms of estradiol. This differential responsiveness is part of the timing mechanism triggering the LH surge.
Collapse
Affiliation(s)
- Paul Micevych
- Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine, Brain Research Institute, University of CaliforniaLos Angeles, CA, USA
- *Correspondence: Paul Micevych, Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1763, USA. e-mail:
| | - Kevin Sinchak
- Department of Biological Sciences, California State UniversityLong Beach, CA, USA
| |
Collapse
|
16
|
Azcoitia I, Santos-Galindo M, Arevalo MA, Garcia-Segura LM. Role of astroglia in the neuroplastic and neuroprotective actions of estradiol. Eur J Neurosci 2010; 32:1995-2002. [DOI: 10.1111/j.1460-9568.2010.07516.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
17
|
|
18
|
Membrane estrogen receptors stimulate intracellular calcium release and progesterone synthesis in hypothalamic astrocytes. J Neurosci 2010; 30:12950-7. [PMID: 20881113 DOI: 10.1523/jneurosci.1158-10.2010] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
In hypothalamic astrocytes obtained from adult female rats, estradiol rapidly increased free cytoplasmic calcium concentrations ([Ca(2+)](i)) that facilitate progesterone synthesis. The present study demonstrated that estradiol (1 nm) significantly and maximally stimulated progesterone synthesis within 5 min, supporting a rapid, nongenomic mechanism. The group I metabotropic glutamate receptor (mGluR1a) antagonist LY 367385 [(S)-(+)-a-amino-4-carboxy-2-methylbenzeneacetic acid] attenuated both the estradiol-induced [Ca(2+)](i) release and progesterone synthesis. To investigate membrane-associated estrogen receptors (mERs), agonists for ERα, ERβ, STX-activated protein, and GPR30 were compared. The selective ERα agonist propylpyrazole triole (PPT) and STX most closely mimicked the estradiol-induced [Ca(2+)](i) responses, where PPT was more potent but less efficacious than STX. Only high doses (100 nm) of selective ERβ agonist diarylpropionitrile (DPN) and GPR30 agonist G-1 induced estradiol-like [Ca(2+)](i) responses. With the exception of DPN (even at 100 nm), all agonists stimulated progesterone synthesis. The PPT- and STX-induced [Ca(2+)](i) release and progesterone synthesis were blocked by LY 367385. While the G-1-stimulated [Ca(2+)](i) release was blocked by LY 367385, progesterone synthesis was not. Since GPR30 was detected intracellularly but not in the membrane, we interpreted these results to suggest that G-1 could activate mGluR1a on the membrane and GPR30 on the smooth endoplasmic reticulum to release intracellular calcium. Although STX and G-1 maximally stimulated [Ca(2+)](i) release in astrocytes from estrogen receptor-α knock-out (ERKO) mice, estradiol in vivo did not stimulate progesterone synthesis in the ERKO mice. Together, these results indicate that mERα is mainly responsible for the rapid, membrane-initiated estradiol-signaling that leads to progesterone synthesis in hypothalamic astrocytes.
Collapse
|
19
|
Actions of estrogens on glial cells: Implications for neuroprotection. Biochim Biophys Acta Gen Subj 2010; 1800:1106-12. [DOI: 10.1016/j.bbagen.2009.10.002] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 09/29/2009] [Accepted: 10/01/2009] [Indexed: 01/21/2023]
|
20
|
Bruce CC, Zhao C, Franklin RJM. Remyelination - An effective means of neuroprotection. Horm Behav 2010; 57:56-62. [PMID: 19538961 DOI: 10.1016/j.yhbeh.2009.06.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 06/04/2009] [Accepted: 06/09/2009] [Indexed: 10/20/2022]
Abstract
Remyelination following central nervous system (CNS) demyelination restores rapid saltatory conduction of action potentials and contributes to the maintenance of axonal integrity. This robust regenerative phenomenon stands in contrast to the limited repair capacity that is characteristic of CNS neuronal injury. However, despite its efficiency in experimental models and some clinical diseases, remyelination failure becomes an increasingly pronounced feature of the pathology of chronic multiple sclerosis (MS) lesions. Chronic demyelination predisposes axons to atrophy, an irreversible event that is a major pathological correlate of progressive functional decline. This has created a compelling case for developing therapies that promote remyelination: evidence from experimental animal models suggests that hormones may have a beneficial role to play in this regard.
Collapse
|
21
|
Abstract
Estradiol has rapid actions in the CNS that are mediated by membrane estrogen receptors (ERs) and activate cell signaling pathways through interaction with metabotropic glutamate receptors (mGluRs). Membrane-initiated estradiol signaling increases the free cytoplasmic calcium concentration ([Ca(2+)](i)) that stimulates the synthesis of neuroprogesterone in astrocytes. We used surface biotinylation to demonstrate that ERalpha has an extracellular portion. In addition to the full-length ERalpha [apparent molecular weight (MW), 66 kDa], surface biotinylation labeled an ERalpha-immunoreactive protein (MW, approximately 52 kDa) identified by both COOH- and NH(2)-directed antibodies. Estradiol treatment regulated membrane levels of both proteins in parallel: within 5 min, estradiol significantly increased membrane levels of the 66 and 52 kDa ERalpha. Internalization, a measure of membrane receptor activation, was also increased by estradiol with a similar time course. Continuous treatment with estradiol for 24-48 h reduced ERalpha levels, suggesting receptor downregulation. Estradiol also increased mGluR1a trafficking and internalization, consistent with the proposed ERalpha-mGluR1a interaction. Blocking ER with ICI 182,780 or mGluR1a with LY 367385 prevented ERalpha trafficking to and from the membrane. Estradiol-induced [Ca(2+)](i) flux was also significantly increased at the time of peak ERalpha activation/internalization. These results demonstrate that ERalpha is present in the membrane and has an extracellular portion. Furthermore, membrane levels and internalization of ERalpha are regulated by estradiol and mGluR1a ligands. The pattern of trafficking into and out of the membrane suggests that the changing concentration of estradiol during the estrous cycle regulates ERalpha to augment and then terminate membrane-initiated signaling.
Collapse
|
22
|
Noell S, Fallier-Becker P, Deutsch U, Mack AF, Wolburg H. Agrin defines polarized distribution of orthogonal arrays of particles in astrocytes. Cell Tissue Res 2009; 337:185-95. [PMID: 19449033 DOI: 10.1007/s00441-009-0812-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 04/21/2009] [Indexed: 11/25/2022]
Abstract
Accumulating evidence indicates that agrin, a heparan sulphate proteoglycan of the extracellular matrix, plays a role in the organization and maintenance of the blood-brain barrier. This evidence is based on the differential effects of agrin isoforms on the expression and distribution of the water channel protein, aquaporin-4 (AQP4), on the swelling capacity of cultured astrocytes of neonatal mice and on freeze-fracture data revealing an agrin-dependent clustering of orthogonal arrays of particles (OAPs), the structural equivalent of AQP4. Here, we show that the OAP density in agrin-null mice is dramatically decreased in comparison with wild-types, by using quantitative freeze-fracture analysis of astrocytic membranes. In contrast, anti-AQP4 immunohistochemistry has revealed that the immunoreactivity of the superficial astrocytic endfeet of the agrin-null mouse is comparable with that in wild-type mice. Moreover, in vitro, wild-type and agrin-null astrocytes cultured from mouse embryos at embryonic day 19.5 differ neither in AQP4 immunoreactivity, nor in OAP density in freeze-fracture replicas. Analyses of brain tissue samples and cultured astrocytes by reverse transcription with the polymerase chain reaction have not demonstrated any difference in the level of AQP4 mRNA between wild-type astrocytes and astrocytes from agrin-null mice. Furthermore, we have been unable to detect any difference in the swelling capacity between wild-type and agrin-null astrocytes. These results clearly demonstrate, for the first time, that agrin plays a pivotal role for the clustering of OAPs in the endfoot membranes of astrocytes, whereas the mere presence of AQP4 is not sufficient for OAP clustering.
Collapse
Affiliation(s)
- Susan Noell
- Institute of Pathology, University of Tübingen, Germany
| | | | | | | | | |
Collapse
|
23
|
Küppers E, Gleiser C, Brito V, Wachter B, Pauly T, Hirt B, Grissmer S. AQP4 expression in striatal primary cultures is regulated by dopamine - implications for proliferation of astrocytes. Eur J Neurosci 2008; 28:2173-82. [DOI: 10.1111/j.1460-9568.2008.06531.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
24
|
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: 65] [Impact Index Per Article: 3.8] [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
|
25
|
Chen N, Napoli JL. All-trans-retinoic acid stimulates translation and induces spine formation in hippocampal neurons through a membrane-associated RARalpha. FASEB J 2007; 22:236-45. [PMID: 17712061 DOI: 10.1096/fj.07-8739com] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Differentiation and patterning in the developing nervous system require the vitamin A metabolite all-trans-retinoic acid (atRA). Recent data suggest that higher cognitive functions, such as creation of hippocampal memory, also require atRA and its receptors, RAR, through affecting synaptic plasticity. Here we show that within 30 min atRA increased dendritic growth approximately 2-fold, and PSD-95 and synaptophysin puncta intensity approximately 3-fold, in cultured mouse hippocampal neurons, suggesting increased synapse formation. atRA (10 nM) increased ERK1/2 phosphorylation within 10 min. In synaptoneurosomes, atRA rapidly increased phosphorylation of ERK1/2, its target 4E-BP, and p70S6K, and its substrate, ribosome protein S6, indicating activation of MAPK and mammalian target of rapamycin (mTOR). Immunofluorescence revealed intense dendritic expression of RARalpha in the mouse hippocampus and localization of RARalpha on the surfaces of primary cultures of hippocampal neurons, with bright puncta along soma and neurites. Surface biotinylation confirmed the locus of RARalpha expression. Knockdown of RARalpha by shRNA impaired atRA-induced spine formation and abolished dendritic growth. Prolonged atRA stimulation reduced surface/total RARalpha by 43%, suggesting internalization, whereas brain-derived nerve growth factor or bicuculline increased the ratio by approximately 1.8-fold. atRA increased translation in the somatodendritic compartment, similar to brain-derived nerve growth factor. atRA specifically increased dendritic translation and surface expression of the alpha-amino-3-hydroxyl-5-methyl-4-isoxazole propionate receptor (AMPAR) subunit 1 (GluR1), without affecting GluR2. These data provide mechanistic insight into atRA function in the hippocampus and identify a unique membrane-associated RARalpha that mediates rapid induction of neuronal translation by atRA.
Collapse
Affiliation(s)
- Na Chen
- Nutritional Science and Toxicology, University of California, Berkeley, California 94720, USA
| | | |
Collapse
|
26
|
Kipp M, Karakaya S, Pawlak J, Araujo-Wright G, Arnold S, Beyer C. Estrogen and the development and protection of nigrostriatal dopaminergic neurons: concerted action of a multitude of signals, protective molecules, and growth factors. Front Neuroendocrinol 2006; 27:376-90. [PMID: 16949139 DOI: 10.1016/j.yfrne.2006.07.001] [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] [Received: 02/02/2006] [Revised: 07/03/2006] [Accepted: 07/10/2006] [Indexed: 01/03/2023]
Abstract
The nigrostriatal dopamine system comprises the dopaminergic neurons located in the ventral midbrain, their axonal connections to the forebrain, and their direct cellular target cells in the striatal complex, i.e. GABAergic neurons. The major function of the nigrostriatal dopaminergic unit is the coordination and fine tuning of motor functions at the extrapyramidal level. Numerous biologically active factors including different types of growth factors (neurotrophins, members of the TGFbeta family, IGFs) and peptide/steroid hormones have been identified in the past to be implicated in the regulation of developmental aspects of this neural system. Some of these developmentally active determinants have in addition been found to play a crucial role in the mediation of neuroprotection concerning dopaminergic neurons. Estrogen was identified as such a compound interfering with embryonic neuronal differentiation and cell survival. The physiological mechanisms underlying these effects are very complex and include interactions with other developmental signals (growth factors), inflammatory processes as well as apoptotic events, but also require the activation of nonneuronal cells such as astrocytes. It appears that estrogen is assuming control over or at least influences a multitude of developmental and protective cellular mechanisms rather than taking over the part of a singular protagonist.
Collapse
Affiliation(s)
- Markus Kipp
- Institute of Neuroanatomy, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Hormonal and locally produced steroids act in the nervous system as neuroendocrine regulators, as trophic factors and as neuromodulators and have a major impact on neural development and function. Glial cells play a prominent role in the local production of steroids and in the mediation of steroid effects on neurons and other glial cells. In this review, we examine the role of glia in the synthesis and metabolism of steroids and the functional implications of glial steroidogenesis. We analyze the mechanisms of steroid signaling on glia, including the role of nuclear receptors and the mechanisms of membrane and cytoplasmic signaling mediated by changes in intracellular calcium levels and activation of signaling kinases. Effects of steroids on functional parameters of glia, such as proliferation, myelin formation, metabolism, cytoskeletal reorganization, and gliosis are also reviewed, as well as the implications of steroid actions on glia for the regulation of synaptic function and connectivity, the regulation of neuroendocrine events, and the response of neural tissue to injury.
Collapse
|
28
|
Mendez P, Cardona-Gomez GP, Garcia-Segura LM. Interactions of insulin-like growth factor-I and estrogen in the brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2005; 567:285-303. [PMID: 16370144 DOI: 10.1007/0-387-26274-1_12] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
29
|
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: 7.3] [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
|
30
|
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: 15] [Impact Index Per Article: 0.8] [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
|
31
|
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: 170] [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
|
32
|
Zhao X, MacBride MM, Peterson BR, Pfaff DW, Vasudevan N. Calcium flux in neuroblastoma cells is a coupling mechanism between non-genomic and genomic modes of estrogens. Neuroendocrinology 2005; 81:174-82. [PMID: 16020926 DOI: 10.1159/000087000] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2004] [Accepted: 03/19/2005] [Indexed: 11/19/2022]
Abstract
Estrogens have been demonstrated to rapidly modulate calcium levels in a variety of cell types. However, the significance of estrogen-mediated calcium flux in neuronal cells is largely unknown. The relative importance of intra- and extracellular sources of calcium in estrogenic effects on neurons is also not well understood. Previously, we have demonstrated that membrane-limited estrogens, such as E-BSA given before an administration of a 2-hour pulse of 17beta-estradiol (E2), can potentiate the transcription mediated by E2 from a consensus estrogen response element (ERE)-driven reporter gene. Inhibitors to signal transduction cascades given along with E-BSA or E2 demonstrated that calcium flux is important for E-BSA-mediated potentiation of transcription in a transiently transfected neuroblastoma cell line. In this report, we have used inhibitors to different voltage-gated calcium channels (VGCCs) and to intracellular store receptors along with E-BSA in the first pulse or with E2 in the second pulse to investigate the relative importance of these channels to estrogen-mediated transcription. Neither L- nor P-type VGCCs seem to play a role in estrogen action in these cells; while N-type VGCCs are important in both the non-genomic and genomic modes of estrogen action. Specific inhibitors also showed that the ryanodine receptor and the inositol trisphosphate receptor are important to E-BSA-mediated transcriptional potentiation. This report provides evidence that while intracellular stores of calcium are required to couple non-genomic actions of estrogen initiated at the membrane to transcription in the nucleus, extracellular sources of calcium are also important in both non-genomic and genomic actions of estrogens.
Collapse
Affiliation(s)
- Xing Zhao
- Department of Biology, Pennsylvania State University, University Park, PA 16802 , USA
| | | | | | | | | |
Collapse
|
33
|
Azcoitia I, Sierra A, Veiga S, Garcia-Segura LM. Aromatase expression by reactive astroglia is neuroprotective. Ann N Y Acad Sci 2004; 1007:298-305. [PMID: 14993062 DOI: 10.1196/annals.1286.028] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The enzyme aromatase catalyzes the conversion of testosterone and other C19 steroids to estradiol. Under normal circumstances, the expression of aromatase in the central nervous system of mammals is restricted to neurons. However, the expression of the enzyme is induced in astrocytes in vitro by stressful stimuli. Furthermore, different types of brain injury induce in vivo the expression of aromatase in reactive astrocytes. The expression of aromatase by reactive astrocytes is neuroprotective, because the pharmacological inhibition of the enzyme in the brain exacerbates neuronal death after different forms of mild neurodegenerative stimuli that do not significantly affect neuronal survival under control conditions. These findings indicate that the induction of aromatase in reactive astrocytes, and the consecutive increase in the local production of estradiol in the brain at injured sites, may be an endogenous neural response to reduce the extent of neurodegenerative damage.
Collapse
Affiliation(s)
- Inigo Azcoitia
- Departamento de Biologïa Celular, Facultad de Biología, Universidad Complutense, E-28040 Madrid, Spain.
| | | | | | | |
Collapse
|
34
|
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
|
35
|
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
|
36
|
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: 95] [Impact Index Per Article: 4.3] [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
|
37
|
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: 39] [Impact Index Per Article: 1.8] [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
|
38
|
Beyer C, Damm N, Brito V, Küppers E. Developmental expression of progesterone receptor isoforms in the mouse midbrain. Neuroreport 2002; 13:877-80. [PMID: 11997705 DOI: 10.1097/00001756-200205070-00028] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Progesterone participates in the regulation of developmental processes in the brain and controls the function of distinct neural circuits. We have studied the expression of progesterone receptor (PR) isoforms in the developing and adult male and female mouse ventral midbrain. Transcripts of both receptor isoforms (PR-A and B) were detectable pre- and postnatally but regulated differentially during ontogeny. Immunoblotting revealed that only the full-length form (PR-B) is transcribed transiently into protein, whereas the truncated PR-A isoform is not detectable as protein. Although the precise function of progesterone in the developing CNS is not fully understood, our data implicate a potential role for PR signaling for the developing nigrostriatal system.
Collapse
Affiliation(s)
- Cordian Beyer
- Anatomie und Zellbiologie, Universität Ulm, 89061 Ulm, Germany
| | | | | | | |
Collapse
|
39
|
Creutz LM, Kritzer MF. Estrogen receptor-beta immunoreactivity in the midbrain of adult rats: regional, subregional, and cellular localization in the A10, A9, and A8 dopamine cell groups. J Comp Neurol 2002; 446:288-300. [PMID: 11932944 DOI: 10.1002/cne.10207] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Estrogen modulates dopamine synthesis, release, and metabolism in corticolimbic and striatal targets of midbrain dopamine neurons. The relevant sites of receptor-mediated action, however, had been elusive, because all available evidence suggested a paucity of intracellular estrogen receptors in the A8, A9, and A10 dopamine regions and their afferent targets. More recent evidence of a relative abundance of the beta isoform of the estrogen receptor (ER) in the substantia nigra and ventral tegmental area (VTA), however, suggests that this newly described receptor may be important in estrogen's stimulation of midbrain DA systems. It is unknown, however, precisely how ERbeta is distributed with respect to the functionally and neurochemically diverse cell populations of the ventral midbrain. To address these issues, this study used single- and double-label immunocytochemistry to detail the regional, subregional, and cellular distributions of ERbeta immunoreactivity in and around midbrain dopamine-containing cell groups in hormonally intact adult male and female rats. These analyses revealed that ERbeta-immunoreactive nuclei were found only in neurons, more specifically, within subsets of both dopaminergic and nondopaminergic neurons in the dorsal VTA, the parabrachial pigmented nucleus, the substantia nigra pars lateralis, the retrorubral fields, and to a lesser extent the linear midline nuclei. These regional and cellular receptor distributions thus place the ERbeta isoform in anatomical register with midbrain dopamine systems known to participate in a spectrum of motor, cognitive, and affective functions.
Collapse
Affiliation(s)
- Lela M Creutz
- Graduate Program in Neurobiology and Behavior, State University of New York at Stony Brook, Stony Brook, NY 11794-5230, USA.
| | | |
Collapse
|
40
|
Ivanova T, Mendez P, Garcia-Segura LM, Beyer C. Rapid stimulation of the PI3-kinase/Akt signalling pathway in developing midbrain neurones by oestrogen. J Neuroendocrinol 2002; 14:73-9. [PMID: 11903815 DOI: 10.1046/j.0007-1331.2001.00742.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Oestrogen promotes the differentiation of neurones in the central nervous system. In the rodent midbrain, the maturation of dopaminergic neurones appears to be under oestrogen control. This is supported by the fact that dopaminergic cells contain nuclear oestrogen receptors-alpha/beta (ER). Second, aromatase is transiently expressed in the developing midbrain. In previous studies, we have shown that oestrogen increases dopamine synthesis and plasticity of dopamine cells. These effects are transmitted through classical nuclear ER but require also the stimulation of nonclassical signalling pathways involving the activation of membrane receptors. This study attempted to identify nonclassical oestrogen-dependent signalling cascades which might be stimulated downstream of membrane ERs. Using cultured mouse midbrain cells, we could demonstrate by Western blotting, that oestrogen rapidly phosphorylates Akt, a kinase which is implicated in the phosphatidylinositol 3 (PI3)-kinase pathway. This effect was only seen in midbrain neurones but not astrocytes. Oestrogen-induced Akt phosphorylation was time- and dose-dependent, showing highest responses after 30 min and at a steroid concentration of 10(-8) and 10(-6) M. Immunocytochemistry for phosphorylated Akt (pAkt) demonstrated that pAkt is predominantly found in a nuclear/perinuclear position and that oestrogen exposure increased the number of pAkt-positive cells. To investigate the mechanisms which are involved in transmitting oestrogen effects on the cellular level, cells were treated with antagonists for distinct signalling pathways. The application of the nuclear ER antagonist ICI 182 780 did not abolish the oestrogen-induced Akt phosphorylation. In contrast, interrupting intracellular calcium signalling with BAPTA completely prevented this effect. The PI3-kinase inhibitor LY294002 also inhibited the activation of Akt by oestrogen. Our study clearly indicates that oestrogen can rapidly stimulate the PI3-kinase/Akt signalling cascade in differentiating midbrain neurones. This effect requires the intermediate activation of calcium-dependent signalling pathways. In conclusion, oestrogen effects in the developing midbrain appear to be connected with the PI3-kinase/Akt signalling mechanism.
Collapse
Affiliation(s)
- T Ivanova
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Ulm, Germany
| | | | | | | |
Collapse
|