401
|
Sasahara K, Shikimi H, Haraguchi S, Sakamoto H, Honda SI, Harada N, Tsutsui K. Mode of action and functional significance of estrogen-inducing dendritic growth, spinogenesis, and synaptogenesis in the developing Purkinje cell. J Neurosci 2007; 27:7408-17. [PMID: 17626201 PMCID: PMC6672615 DOI: 10.1523/jneurosci.0710-07.2007] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Revised: 05/30/2007] [Accepted: 06/02/2007] [Indexed: 11/21/2022] Open
Abstract
Neurosteroids are synthesized de novo from cholesterol in the brain. To understand neurosteroid action in the brain, data on the regio- and temporal-specific synthesis of neurosteroids are needed. Recently, we identified the Purkinje cell as an active neurosteroidogenic cell. In rodents, this neuron actively produces several neurosteroids including estradiol during neonatal life, when cerebellar neuronal circuit formation occurs. Estradiol may be involved in cerebellar neuronal circuit formation through promoting neuronal growth and neuronal synaptic contact, because the Purkinje cell expresses estrogen receptor-beta (ERbeta). To test this hypothesis, in this study we examined the effects of estradiol on dendritic growth, spinogenesis, and synaptogenesis in the Purkinje cell using neonatal wild-type (WT) mice or cytochrome P450 aromatase knock-out (ArKO) mice. Administration of estradiol to neonatal WT or ArKO mice increased dendritic growth, spinogenesis, and synaptogenesis in the Purkinje cell. In contrast, WT mice treated with tamoxifen, an ER antagonist, or ArKO mice exhibited decreased Purkinje dendritic growth, spinogenesis, and synaptogenesis at the same neonatal period. To elucidate the mode of action of estradiol, we further examined the expression of brain-derived neurotrophic factor (BDNF) in response to estrogen actions in the neonate. Estrogen administration to neonatal WT or ArKO mice increased the BDNF level in the cerebellum, whereas tamoxifen decreased the BDNF level in WT mice similar to ArKO mice. BDNF administration to tamoxifen-treated WT mice increased Purkinje dendritic growth. These results indicate that estradiol induces dendritic growth, spinogenesis, and synaptogenesis in the developing Purkinje cell via BDNF action during neonatal life.
Collapse
Affiliation(s)
- Katsunori Sasahara
- Laboratory of Brain Science, Faculty of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Hanako Shikimi
- Laboratory of Brain Science, Faculty of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Shogo Haraguchi
- Laboratory of Brain Science, Faculty of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
- Laboratory of Integrative Brain Sciences, Department of Biology, Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo 169-8050, Japan
| | - Hirotaka Sakamoto
- Laboratory of Brain Science, Faculty of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Shin-ichiro Honda
- Department of Biochemistry, School of Medicine, Fujita Health University, Aichi 470-1192, Japan, and
| | - Nobuhiro Harada
- Department of Biochemistry, School of Medicine, Fujita Health University, Aichi 470-1192, Japan, and
| | - Kazuyoshi Tsutsui
- Laboratory of Brain Science, Faculty of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
- Laboratory of Integrative Brain Sciences, Department of Biology, Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo 169-8050, Japan
| |
Collapse
|
402
|
Grassi S, Frondaroli A, Dieni C, Dutia MB, Pettorossi VE. Neurosteroid modulation of neuronal excitability and synaptic transmission in the rat medial vestibular nuclei. Eur J Neurosci 2007; 26:23-32. [PMID: 17596193 DOI: 10.1111/j.1460-9568.2007.05645.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
In rat brainstem slices, we investigated the influence of the neurosteroids tetrahydrodeoxycorticosterone (THDOC) and allopregnanolone (ALLO) on the synaptically driven and spontaneous activity of vestibular neurons, by analysing their effects on the amplitude of the field potentials evoked in the medial vestibular nuclei (MVN) by vestibular afferent stimulation and on the spontaneous firing rate of MVN neurons. Furthermore, the interaction with gamma-aminobutyric acid (GABA) and glutamate receptors was analysed by using specific antagonists for GABA(A) (bicuculline), alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/ kainate [2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo(f)quinoxaline-7-sulphonamide disodium salt (NBQX)], N-methyl-D-aspartate (NMDA) [D-(-)-2-amino-5-phosphonopentanoic acid (AP-5)] and group I metabotropic glutamate receptors (mGlu-I) [(R,S)-1-aminoindan-1,5-dicarboxylic acid (AIDA)] receptors. THDOC and ALLO evoked two opposite long-lasting effects, consisting of either a potentiation or a reduction of field potential and firing rate, which showed early and late components, occurring in conjunction or separately after neurosteroid application. The depressions depended on GABA(A) receptors, as they were abolished by bicuculline, while early potentiation involved glutamate AMPA/kainate receptors, as NBQX markedly reduced the incidence of early firing rate enhancement and, in the case of ALLO, even provoked depression. This suggests that THDOC and ALLO enhance the GABA(A) inhibitory influence on the MVN neurons and facilitate the AMPA/kainate facilitatory one. Conversely, a late potentiation effect, which was still induced after glutamate and GABA(A) receptor blockade, might involve a different mechanism. We conclude that the modulation of neuronal activity in the MVN by THDOC and ALLO, through their actions on GABA(A) and AMPA/kainate receptors, may have a physiological role in regulating the vestibular system function under normal conditions and during the stress response that accompanies many forms of vestibular dysfunction.
Collapse
Affiliation(s)
- Silvarosa Grassi
- Department of Internal Medicine, Section of Human Physiology, University of Perugia, Via del Giochetto, I-06126 Perugia, Italy.
| | | | | | | | | |
Collapse
|
403
|
Rego JLD, Leprince J, Luu-The V, Pelletier G, Tonon MC, Vaudry H. Structure−Activity Relationships of a Series of Analogs of the Endozepine Octadecaneuropeptide (ODN11-18) on Neurosteroid Biosynthesis by Hypothalamic Explants. J Med Chem 2007; 50:3070-6. [PMID: 17550241 DOI: 10.1021/jm0610548] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have previously shown that the endozepine octadecaneuropeptide (ODN) stimulates the biosynthesis of neurosteroids from frog hypothalamic explants. In the present study, we have investigated the structure-activity relationships of a series of analogs of the C-terminal octapeptide of ODN (OP) on neurosteroid formation. We found that OP and its cyclic analog cyclo1-8OP stimulate in a concentration-dependent manner the synthesis of various steroids including 17-hydroxypregnenolone, progesterone, 17-hydroxyprogesterone and dehydroepiandrosterone. Deletion or Ala-substitution of the Arg1 or Pro2 residues of OP did not affect the activity of the peptide. In contrast, deletion or replacement of any of the amino acids of the C-terminal hexapeptide fragment totally abolished the effect of OP on neurosteroid biosynthesis. The present study indicates that the C-terminal hexapeptide of ODN/OP is the minimal sequence retaining full biological activity on steroid-producing neurons.
Collapse
Affiliation(s)
- Jean Luc Do Rego
- INSERM U413, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research (IFRMP 23), University of Rouen, 76821 Mont-Saint-Aignan, France
| | | | | | | | | | | |
Collapse
|
404
|
Verdi J, Ahmadiani A. Finasteride, a 5alpha-reductase inhibitor, potentiates antinociceptive effects of morphine, prevents the development of morphine tolerance and attenuates abstinence behavior in the rat. Horm Behav 2007; 51:605-10. [PMID: 17428486 DOI: 10.1016/j.yhbeh.2007.02.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Revised: 02/10/2007] [Accepted: 02/22/2007] [Indexed: 11/29/2022]
Abstract
It has been shown that morphine increases 5alpha-reductase enzyme activity in the rat central nervous system; however importance of this finding on morphine analgesia, tolerance and dependence has not been reported. In the present study, we investigated inhibition of 5alpha-reductase enzyme on morphine effects using finasteride. To determine whether the 5alpha-reductase enzyme interact with morphine analgesia, finasteride (5 mg/kg, i.p.) was administrated with morphine (5 and 7 mg/kg, i.p.). The tail-flick test was used to assess the nociceptive threshold, before and 15, 30, 45, 60 and 90 min after drug administration. In tolerance experiments, morphine 20 mg/kg was injected i.p., twice daily for 4 days. The development and expression of dependence were assessed in the naloxone precipitation test 5 days after the morphine (20-30 mg/kg, i.p.) administration. We found that finasteride could potentiate the antinociceptive effect of morphine. In addition, chronic finasteride administration effectively blocked development of tolerance and dependence to morphine. Following chronic morphine administration, single dose injection of finasteride failed to reverse tolerance but prevented naloxone precipitate withdrawal syndrome. Therefore, it was concluded that there is a functional relationship between 5alpha-reductase enzyme and morphine.
Collapse
Affiliation(s)
- Javad Verdi
- Department of Physiology and Pharmacology, Kashan University of Medical Sciences, Kashan, Iran
| | | |
Collapse
|
405
|
Johansson T, Elfverson M, Birgner C, Frändberg PA, Nyberg F, Le Grevès P. Neurosteroids alter glutamate-induced changes in neurite morphology of NG108-15 cells. J Steroid Biochem Mol Biol 2007; 104:215-9. [PMID: 17512193 DOI: 10.1016/j.jsbmb.2007.03.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Activation of the NMDA receptor leads to increased intracellular Ca2+ levels ([Ca2+]i) which induces outgrowth of and morphologic changes in the neurites of the NG108-15 cell line. This effect can be blocked by antagonists for this glutamate receptor subtype (e.g. ifenprodil or AP5). We have previously shown that nanomolar concentrations of various neurosteroids modulate ifenprodil binding to the NMDA receptor. To investigate whether this interaction affects the functioning of the receptor, we studied the effect of 24 and 48 h of pregnenolone sulphate (PS) or pregnanolone sulphate (3alpha5betaS) on glutamate-stimulated NG108-15 cells. Unexpectedly, the neurosteroids themselves had an inhibitory effect on glutamate-induced changes in neurite patterns. This effect was comparable to that of ifenprodil or AP5. Moreover, the effect of combined treatment with 3alpha5betaS and ifenprodil on neurite morphology indicated a functional interaction between the substances. Interestingly, PS induced cell detachment over time, an effect that was further enhanced by ifenprodil. Cell detachment was also seen after 48 h of treatment with 3alpha5betaS; however, the effect was blocked by ifenprodil and weaker than that of PS. The interaction with the NR2B-selective antagonist ifenprodil indicates that this NMDA receptor subunit may be involved in neurosteroid-induced NG108-15 cell detachment.
Collapse
Affiliation(s)
- Tobias Johansson
- Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, S-751 24 Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
406
|
Lyssimachou A, Arukwe A. Alteration of brain and interrenal StAR protein, P450scc, and Cyp11beta mRNA levels in atlantic salmon after nominal waterborne exposure to the synthetic pharmaceutical estrogen ethynylestradiol. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2007; 70:606-13. [PMID: 17365614 DOI: 10.1080/10937400600882905] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Pharmaceuticals are ubiquitous pollutants in the aquatic environment, where their potential effects on nontarget species like fish has only recently become subject of systematic investigations. Recently, it was shown that the documented xenoestrogen nonylphenol produced variations in brain steroidogenic acute regulatory (StAR) protein, cytochrome P-450-mediated cholesterol side-chain cleavage (P450scc), and cytochrome P-45011beta hydroxylase (CYP11beta) gene transcripts of exposed juvenile salmon (Arukwe, 2005). In the present study, experiments were undertaken to examine the effect of the synthetic pharmaceutical endocrine disruptor ethynylestradiol (EE2), given in water at 5 or 50 ng/L and sampled at d 0 (control), 3, and 7 after exposure, on these key and rate-limiting brain and interrenal steroidogenic pathways of juvenile salmon using quantitative (real-time) polymerase chain reaction (qPCR). Our data, which are based on nominal exposure concentrations, show that brain and head kidney StAR and P450scc expression were modulated by EE2 in a time- and concentration-specific manner. While the StAR protein and P450scc showed EE2 concentration-dependent transcriptional increases in the brain and head kidney at d 3 after exposure, no significant effect was observed at d 7. The EE2 induced effects at d 7 were underscored because the carrier solvent (dimethyl sulfoxide, DMSO) produced significant induction of the StAR protein and P450scc in both the brain and head kidney at d 7 compared to d 3 postexposure. CYP11beta transcript was detected in the brain and head kidney, where the expression patterns were modulated by EE2 in a concentration-and time-specific manner. In the brain, DMSO produced significant changes in the CYP11beta gene expression at d 7 compared to d 3 after exposure. These changes in the levels of StAR, P450scc, and CYP11beta mRNA levels in important steroidogenic organs suggest that the experimental animals are experiencing a time-dependent impaired steroidogenesis. Thus, the StAR protein, P450scc, and CYP11beta might represent sensitive diagnostic tools for short-term and acute exposure to endocrine disrupting chemicals. In view of the present study and high concentrations of EE2 reported in effluents and surface waters from Europe and the United States, pharmaceuticals in the environment represent potentially more serious health concern both to humans and wildlife than earlier anticipated.
Collapse
Affiliation(s)
- Angeliki Lyssimachou
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | | |
Collapse
|
407
|
Charalampopoulos I, Alexaki VI, Tsatsanis C, Minas V, Dermitzaki E, Lasaridis I, Vardouli L, Stournaras C, Margioris AN, Castanas E, Gravanis A. Neurosteroids as endogenous inhibitors of neuronal cell apoptosis in aging. Ann N Y Acad Sci 2007; 1088:139-52. [PMID: 17192562 DOI: 10.1196/annals.1366.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The neuroactive steroids dehydroepiandrosterone (DHEA), its sulfate ester DHEAS, and allopregnanolone (Allo) are produced in the adrenals and the brain. Their production rate and levels in serum, brain, and adrenals decrease gradually with advancing age. The decline of their levels was associated with age-related neuronal dysfunction and degeneration, most probably because these steroids protect central nervous system (CNS) neurons against noxious agents. Indeed, DHEA(S) protects rat hippocampal neurons against NMDA-induced excitotoxicity, whereas Allo ameliorates NMDA-induced excitotoxicity in human neurons. These steroids exert also a protective role on the sympathetic nervous system. Indeed, DHEA, DHEAS, and Allo protect chromaffin cells and the sympathoadrenal PC12 cells (an established model for the study of neuronal cell apoptosis and survival) against serum deprivation-induced apoptosis. Their effects are time- and dose-dependent with EC(50) 1.8, 1.1, and 1.5 nM, respectively. The prosurvival effect of DHEA(S) appears to be NMDA-, GABA(A)- sigma1-, or estrogen receptor-independent, and is mediated by G-protein-coupled-specific membrane binding sites. It involves the antiapoptotic Bcl-2 proteins, and the activation of prosurvival transcription factors CREB and NF-kappaB, upstream effectors of the antiapoptotic Bcl-2 protein expression, as well as prosurvival kinase PKCalpha/beta, a posttranslational activator of Bcl-2. Furthermore, they directly stimulate biosynthesis and release of neuroprotective catecholamines, exerting a direct transcriptional effect on tyrosine hydroxylase, and regulating actin depolymerization and submembrane actin filament disassembly, a fast-response cellular system regulating trafficking of catecholamine vesicles. These findings suggest that neurosteroids may act as endogenous neuroprotective factors. The decline of neurosteroid levels during aging may leave the brain unprotected against neurotoxic challenges.
Collapse
|
408
|
Dong LY, Cheng ZX, Fu YM, Wang ZM, Zhu YH, Sun JL, Dong Y, Zheng P. Neurosteroid dehydroepiandrosterone sulfate enhances spontaneous glutamate release in rat prelimbic cortex through activation of dopamine D1 and sigma-1 receptor. Neuropharmacology 2007; 52:966-74. [PMID: 17141281 DOI: 10.1016/j.neuropharm.2006.10.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 10/12/2006] [Accepted: 10/20/2006] [Indexed: 11/30/2022]
Abstract
This paper studied the effect of neurosteroid dehydroepiandrosterone sulfate on spontaneous glutamate release in the prelimbic cortex by using electrophysiological and biochemical methods combined with a pharmacological approach and made some comparisons with those in the hippocampus. The results showed that dehydroepiandrosterone sulfate increased the frequency of miniature excitatory postsynaptic currents in the prelimbic cortex and hippocampus; sigma-1 receptor antagonist partially blocked the effect of dehydroepiandrosterone sulfate in the prelimbic cortex, but completely blocked it in the hippocampus; D1 receptor antagonist, adenylyl cyclase inhibitor and protein kinase A inhibitor completely blocked the effect of dehydroepiandrosterone sulfate in the prelimbic cortex; dehydroepiandrosterone sulfate increased the activity of protein kinase A in the prelimbic cortex and hippocampus; the effect of dehydroepiandrosterone sulfate on protein kinase A was completely blocked by sigma-1 receptor antagonist in the hippocampus, but was partially blocked in the prelimbic cortex; interestingly, here again, the effect of dehydroepiandrosterone sulfate on protein kinase A was completely blocked by D1 receptor antagonist in the prelimbic cortex. These results suggest that dehydroepiandrosterone sulfate promotes presynaptic glutamate release in the prelimbic cortex via activation of D1 and sigma-1 receptors.
Collapse
Affiliation(s)
- Lian-Yan Dong
- State Key Laboratory of Medical Neurobiology, Fudan University Shanghai Medical College, 138 Yixueyuan Road, Shanghai 200032, PR China
| | | | | | | | | | | | | | | |
Collapse
|
409
|
Steroid modulation of GABAA receptor-mediated transmission in the hypothalamus: effects on reproductive function. Neuropharmacology 2007; 52:1439-53. [PMID: 17433821 DOI: 10.1016/j.neuropharm.2007.01.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 01/30/2007] [Accepted: 01/31/2007] [Indexed: 10/23/2022]
Abstract
The hypothalamus, the seat of neuroendocrine control, is exquisitely sensitive to gonadal steroids. For decades it has been known that androgens, estrogens and progestins, acting through nuclear hormone receptors, elicit both organizational and activational effects in the hypothalamus and basal forebrain that are essential for reproductive function. While changes in gene expression mediated by these classical hormone pathways are paramount in governing both sexual differentiation and the neural control of reproduction, it is also clear that steroids impart critical control of neuroendocrine functions through non-genomic mechanisms. Specifically, endogenous neurosteroid derivatives of deoxycorticosterone, progesterone and testosterone, as well and synthetic anabolic androgenic steroids that are self-administered as drugs of abuse, elicit acute effects via allosteric modulation of gamma-aminobutyric acid type A receptors. GABAergic transmission within the hypothalamus and basal forebrain is a key regulator of pubertal onset, the expression of sexual behaviors, pregnancy and parturition. Summarized here are the known actions of steroid modulators on GABAergic transmission within the hypothalamus/basal forebrain, with a focus on the medial preoptic area and the supraoptic/paraventricular nuclei that are known to be central players in the control of reproduction.
Collapse
|
410
|
Cascio C, Russo D, Drago G, Galizzi G, Passantino R, Guarneri R, Guarneri P. 17beta-estradiol synthesis in the adult male rat retina. Exp Eye Res 2007; 85:166-72. [PMID: 17466975 DOI: 10.1016/j.exer.2007.02.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Revised: 01/31/2007] [Accepted: 02/04/2007] [Indexed: 11/21/2022]
Abstract
17beta-Estradiol (E2) exerts neurotrophic and neuroprotective effects in the retina as well as in other CNS structures, independently of sex. Retinal effects, however, have not been supported by evidence on local synthesis, and whether CNS 17beta-estradiol is formed in a neurosteroidogenic pathway starting from cholesterol conversion into pregnenolone is a question still left unanswered. In the adult male rat retina, we have previously showed localization and activity of the P450 side chain cleavage (P450scc) enzyme, which is involved in pregnenolone synthesis. Here, we demonstrate both the mRNA and protein expression of 3beta-hydroxysteroid dehydrogenase (3beta-HSD), P450aromatase and also of P450scc, but only the protein expression of P450 17alpha-hydroxylase/lyase (P450c17). Using radiolabeled pregnenolone and testosterone as precursors, in the isolated and intact retina of adult male rats, E2 is produced in a large amount by each precursor within 1-4h, suggesting a highly active metabolic pathway towards its formation. The immunolocalization pattern shows enzymes and estrogen receptor subtypes (ERalpha, ERbeta) scattered in the retina with different intensities throughout the layers. The results point to the adult male rat retina as a neurosteroidogenic structure where E2 synthesis via a progesterone pathway and the presence of estrogen receptors provide important clues for understanding the neurotrophic and neuroprotective effects of the steroid hormone.
Collapse
Affiliation(s)
- C Cascio
- Istituto di Biomedicina e Immunologia Molecolare, IBIM - CNR, Via Ugo La Malfa, 153, 90146 Palermo, Italy
| | | | | | | | | | | | | |
Collapse
|
411
|
Zhou L, Lehan N, Wehrenberg U, Disteldorf E, von Lossow R, Mares U, Jarry H, Rune GM. Neuroprotection by estradiol: A role of aromatase against spine synapse loss after blockade of GABAA receptors. Exp Neurol 2007; 203:72-81. [PMID: 17005180 DOI: 10.1016/j.expneurol.2006.07.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2006] [Revised: 07/19/2006] [Accepted: 07/24/2006] [Indexed: 11/24/2022]
Abstract
Estrogen has been suggested to be pro-epileptic by reducing GABA synthesis, resulting in increased spine density and a decreased threshold for seizures in the hippocampus, which, once they occur, are characterized by a dramatic spine loss in the affected brain areas. As considerable amounts of estradiol are synthesized in the hippocampus, in this study we focused on aromatase, the rate-limiting enzyme in estrogen synthesis in order to examine the role of locally synthesized estrogens in epilepsy. To this end, we first examined the effects of letrozole, a potent aromatase inhibitor, on GABA metabolism in single interneurons of hippocampal dispersion cultures. Letrozole downregulated estradiol release into the medium, as well as glutamate decarboxylase (GAD) expression and GABA synthesis, and decreased the number of GAD positive cells in the cultures. Next, we counted spine synapses and measured estradiol release of hippocampal slice cultures, in which GABA(A) receptors had been blocked by bicuculline, in order to mimic epileptic activity. Treatment of slice cultures with bicuculline resulted in a dramatic decrease in the number of spine synapses and in a significant suppression of estrogen synthesis. The decrease in synapse number in response to bicuculline was restored by combined application of estradiol and bicuculline. Surprisingly, estradiol alone had no effect on either spine synapse number or on GAD expression and GABA synthesis. "Rescue" of synapse number in "epileptic slices" by estradiol and maintenance of GABA metabolism by hippocampus-derived estradiol points to a neuroprotective role of aromatase in epilepsy. Re-filling of estradiol stores after their depletion due to overexcitation may therefore add to therapeutical strategies in epilepsy.
Collapse
Affiliation(s)
- Lepu Zhou
- Institute of Anatomy I, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246 Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
412
|
Westholm DE, Rumbley JN, Salo DR, Rich TP, Anderson GW. Organic anion-transporting polypeptides at the blood-brain and blood-cerebrospinal fluid barriers. Curr Top Dev Biol 2007; 80:135-70. [PMID: 17950374 DOI: 10.1016/s0070-2153(07)80004-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organic anion-transporting polypeptides (Oatps) are solute carrier family members that exhibit marked evolutionary conservation. Mammalian Oatps exhibit wide tissue expression with an emphasis on expression in barrier cells. In the brain, Oatps are expressed in the blood-brain barrier endothelial cells and blood-cerebrospinal fluid barrier epithelial cells. This expression profile serves to illustrate a central role for Oatps in transporting endo- and xenobiotics across brain barrier cells. This chapter will detail the expression patterns and substrate specificities of Oatps expressed in the brain, and will place special emphases on the role of Oatps in prostaglandin synthesis and in the transport of conjugated endobiotics.
Collapse
Affiliation(s)
- Daniel E Westholm
- College of Pharmacy, University of Minnesota, Duluth, Minnesota 55812, USA
| | | | | | | | | |
Collapse
|
413
|
London SE, Schlinger BA. Steroidogenic enzymes along the ventricular proliferative zone in the developing songbird brain. J Comp Neurol 2007; 502:507-21. [PMID: 17394140 DOI: 10.1002/cne.21335] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neural development requires regulation and coordination of the differentiation, migration, and survival of newly divided cells, most of which derive from the region surrounding the lateral ventricles. While many factors are involved in these maturational processes, studies of cell proliferation and neurogenesis in songbirds indicate that sex steroids may provide crucial cues to newly divided cells and may be fundamental to the organization of a specific neural circuit, the song system. In the case of the zebra finch, steroids that impact song system masculinization are most likely not synthesized from the gonads but from the brain, and evidence is mounting that both developing and adult zebra finches have the capacity for neurosteroidogenesis. Therefore, we hypothesized that during early development, all of the genes required for de novo sex steroid synthesis would be expressed in regions that would indicate a role for neurosteroids in neural organization. We found that the genes necessary for de novo neurosteroid synthesis at posthatch day 1 (P1) and P5 show a broad expression distribution. Most strikingly, the spatial distribution of expression for all of the genes necessary for androgen synthesis is similar to the previously described pattern of proliferating neuronal precursors along the lateral border of the lateral ventricle. Due to the increasing evidence for neurosteroid action on multiple cell traits, it may be that locally synthesized neurosteroids impact cells along the proliferative zone to influence early events in neural development generally and song system masculinization specifically.
Collapse
Affiliation(s)
- Sarah E London
- Interdepartmental Program in Neuroscience, Department of Physiological Science, Laboratory of Neuroendocrinology, University of California, Los Angeles, California 90095-1606, USA
| | | |
Collapse
|
414
|
Chang TY, Chang CCY, Ohgami N, Yamauchi Y. Cholesterol sensing, trafficking, and esterification. Annu Rev Cell Dev Biol 2006; 22:129-57. [PMID: 16753029 DOI: 10.1146/annurev.cellbio.22.010305.104656] [Citation(s) in RCA: 455] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mammalian cells acquire cholesterol from low-density lipoprotein (LDL) and from endogenous biosynthesis. The roles of the Niemann-Pick type C1 protein in mediating the endosomal transport of LDL-derived cholesterol and endogenously synthesized cholesterol are discussed. Excess cellular cholesterol is converted to cholesteryl esters by the enzyme acyl-coenzyme A:cholesterol acyltransferase (ACAT) 1 or is removed from a cell by cellular cholesterol efflux at the plasma membrane. A close relationship between the ACAT substrate pool and the cholesterol efflux pool is proposed. Sterol-sensing domains (SSDs) are present in several membrane proteins, including NPC1, HMG-CoA reductase, and the SREBP cleavage-activating protein. The functions of SSDs are described. ACAT1 is an endoplasmic reticulum cholesterol sensor and contains a signature motif characteristic of the membrane-bound acyltransferase family. The nonvesicular cholesterol translocation processes involve the START domain proteins and the oxysterol binding protein-related proteins (ORPs). The properties of these proteins are summarized.
Collapse
Affiliation(s)
- Ta-Yuan Chang
- Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire 03755, USA.
| | | | | | | |
Collapse
|
415
|
Tsutsui K. Biosynthesis, mode of action and functional significance of neurosteroids in the developing Purkinje cell. J Steroid Biochem Mol Biol 2006; 102:187-94. [PMID: 17113981 DOI: 10.1016/j.jsbmb.2006.09.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
New findings over the past decade have shown that the brain has the capability of forming steroids de novo from cholesterol, the so-called "neurosteroids". To understand neurosteroid action in the brain, data on the regio- and temporal-specific synthesis of neurosteroids are needed. Recently, we have demonstrated that the Purkinje cell, a cerebellar neuron, is a major site for neurosteroid formation in a variety of vertebrates. This is the first demonstration of de novo neuronal neurosteroidogenesis in the brain. Since this discovery, organizing actions of neurosteroids are becoming clear by the studies on mammals using the Purkinje cell as an excellent cellular model. In mammals, the Purkinje cell actively synthesizes progesterone de novo from cholesterol during neonatal life, when cerebellar neuronal circuit formation occurs. The Purkinje cell may also produces estradiol in the neonate. Interestingly, both progesterone and estradiol promote dendritic growth, spinogenesis and synaptogenesis via each cognate nuclear receptor in the developing Purkinje cell. Such organizing actions may contribute to the formation of cerebellar neuronal circuit during neonatal life. This paper summarizes the advances made in our understanding of the biosynthesis, mode of action and functional significance of neurosteroids in the developing Purkinje cell.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Brain Science, Faculty of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan.
| |
Collapse
|
416
|
London SE, Monks DA, Wade J, Schlinger BA. Widespread capacity for steroid synthesis in the avian brain and song system. Endocrinology 2006; 147:5975-87. [PMID: 16935847 PMCID: PMC2903432 DOI: 10.1210/en.2006-0154] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Steroids exert powerful effects on the brains and behavior of many species, but measures and manipulations of endocrine physiology in songbirds often reveal unexplained connections between steroids and the brain. The zebra finch song system, a sensorimotor neural circuit sensitive to steroids throughout life, organizes and functions largely in apparent independence from gonadally derived steroids. We tested the hypothesis that the zebra finch brain has the capacity for de novo steroidogenesis and that neurally synthesized steroids, neurosteroids, may impact the song system. Using multiple techniques, we demonstrate that the steroidogenic acute regulatory protein (StAR), cytochrome P450 side-chain cleavage (CYP11A1), and 3beta-hydroxysteroid dehydrogenase/Delta5-Delta4 isomerase, the first three factors in the steroidogenic pathway, are expressed in both developing and adult zebra finch brain. Detailed expression mapping at posthatch d 20 (P20) and adult reveals widespread area-specific expression and coexpression patterns for steroidogenic acute regulatory protein, CYP11A1, and 3beta-hydroxysteroid dehydrogenase/Delta5-Delta4 isomerase, which suggest neurosteroids may modulate multiple brain functions, including sensory and motor systems. Notably, whereas expression of other steroidogenic genes such as aromatase has been essentially absent from the song system, each of the major song nuclei express at least a subset of steroidogenic genes described here, establishing the song system as a potential steroidogenic circuit.
Collapse
Affiliation(s)
- Sarah E London
- University of California, Los Angeles, 621 Charles Young Drive South, P.O. Box 951606, Los Angeles, California 90095-1606, USA.
| | | | | | | |
Collapse
|
417
|
Billiards SS, Nguyen PN, Scheerlinck JP, Phillips DJ, Canny BJ, Walker DW, Hirst JJ. Hypoxia Potentiates Endotoxin-Induced Allopregnanolone Concentrations in the Newborn Brain. Neonatology 2006; 90:258-67. [PMID: 16804294 DOI: 10.1159/000094146] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Accepted: 02/21/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND Allopregnanolone is a neurosteroid produced in the brain that can alter the excitability of the CNS. Neurosteroids have neuroprotective properties, and their elevation in response to stress may protect the newborn brain following infection or hypoxia. Infection, particularly of the respiratory tract, may lead to episodes of hypoxia. Infection and hypoxia have been identified as factors contributing to neonatal morbidity and mortality. OBJECTIVES To determine the effect of acute episodes of hypoxia alone or in combination with lipopolysaccharide (LPS) exposure on plasma and brain allopregnanolone concentrations in lambs 10-21 days old. Also, to examine plasma levels of cortisol and the cytokines, tumour necrosis factor-alpha and interleutkin-6 after these challenges. RESULTS Allopregnanolone concentrations in the brain were markedly increased after hypoxia. Hypoxia following prior LPS treatment resulted in greater increases in brain allopregnanolone concentrations compared to either the LPS or hypoxia treatment alone. Importantly, brain regions unaffected by LPS or hypoxia alone (thalamus/hypothalamus, cerebellum) showed significant increases of allopregnanolone content following the combined LPS and hypoxia treatments. Plasma tumour necrosis factor-alpha and interleukin-6 concentrations were increased after LPS treatment with and without hypoxia, but not by hypoxia alone. In contrast, plasma cortisol concentrations were increased after both stressors. CONCLUSIONS These results show that the brain of young lambs readily responds to physiological stress by increased production of allopregnanolone. This response may protect the developing brain from the cytotoxicity following hypoxic and infectious episodes.
Collapse
|
418
|
Abstract
This review summarizes the mechanisms of cellular cholesterol transport and monogenic human diseases caused by defects in intracellular cholesterol processing. In addition, selected mouse models of disturbed cholesterol trafficking are discussed. Current pharmacological strategies to prevent atherosclerosis are largely based on altering cellular cholesterol balance and are introduced in this context. Finally, because of the organizing potential of cholesterol in membranes, disturbances in cellular cholesterol transport have implications for a wide variety of human diseases, of which selected examples are given.
Collapse
Affiliation(s)
- Elina Ikonen
- Institute of Biomedicine/Anatomy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
419
|
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
|
420
|
Quezada M, Henríquez S, Vargas M, Cardenas H, Tapia A, Rios M, Salvatierra AM, Orihuela PA, Croxatto HB, Velasquez L. Proenkephalin A and the gamma-aminobutyric acid A receptor pi subunit: expression, localization, and dynamic changes in human secretory endometrium. Fertil Steril 2006; 86:1750-7. [PMID: 17074347 DOI: 10.1016/j.fertnstert.2006.05.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2005] [Revised: 05/06/2006] [Accepted: 05/06/2006] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To compare mRNA and protein levels of proenkephalin A (PEA) and gamma-aminobutyric acid A receptor pi subunit (piGABA-R) in human secretory endometrium before and during receptivity and to determine the cell phenotypes where they are expressed. DESIGN Prospective and observational, comparing prereceptive vs. receptive stages of secretory endometrium within the same nonconceptional menstrual cycle. SETTING University and non-governmental organization (NGO)-based academic and clinical-research facilities. PATIENT(S) Seven healthy, multiparous, surgically sterilized women with spontaneous regular menstrual cycles. INTERVENTION(S) Endometrial biopsies were obtained on LH+3 and LH+7 within the same cycle. MAIN OUTCOME MEASURE(S) Levels of PEA and piGABA-R mRNA were determined by real-time PCR, and protein presence, by immunofluorescence. RESULT(S) The mRNA level of PEA fell, whereas that of piGABA-R increased, during endometrial receptivity. Positive immunostaining of PEA was found in the luminal and glandular epithelium, whereas that of piGABA-R was in luminal epithelium and stromal cells. CONCLUSION(S) The discrete cell-phenotype localization and timing of the changes in the level of PEA and of piGABA-R mRNA and protein suggest an important role for these molecules in switching the human endometrium from a refractory to a receptive state.
Collapse
Affiliation(s)
- Marisol Quezada
- Laboratorio de Inmunología de la Reproducción, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | |
Collapse
|
421
|
Rasmusson AM, Pinna G, Paliwal P, Weisman D, Gottschalk C, Charney D, Krystal J, Guidotti A. Decreased cerebrospinal fluid allopregnanolone levels in women with posttraumatic stress disorder. Biol Psychiatry 2006; 60:704-13. [PMID: 16934764 DOI: 10.1016/j.biopsych.2006.03.026] [Citation(s) in RCA: 208] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Revised: 03/03/2006] [Accepted: 03/07/2006] [Indexed: 11/17/2022]
Abstract
BACKGROUND Alterations in the gamma-amino-butyric acid (GABA) neurotransmitter system have been identified in some populations with posttraumatic stress disorder (PTSD). METHODS To further investigate factors of relevance to GABAergic neurotransmission in PTSD, we measured cerebrospinal fluid (CSF) levels of allopregnanolone and pregnanolone combined (ALLO: congeners that potently and positively modulate effects of GABA at the GABA(A) receptor), 5alpha-dihydroprogesterone (5alpha-DHP: the immediate precursor for allopregnanolone), dehydroepiandrosterone (DHEA: a negative modulator of GABA(A) receptor function), and progesterone with gas chromatography, mass spectrometry in premenopausal women with (n = 9) and without (n = 10) PTSD. Subjects were free of psychotropic medications, alcohol, and illicit drugs; all were in the follicular phase of the menstrual cycle except three healthy and four PTSD subjects receiving oral contraceptives. RESULTS There were no group differences in progesterone, 5alpha-DHP, or DHEA levels. The PTSD group ALLO levels were < 39% of healthy group levels. The ALLO/DHEA ratio correlated negatively with PTSD re-experiencing symptoms (n = -.82, p < 008; trend) and with Profile of Mood State depression/dejection scores (n = -0.70, p < 0008). CONCLUSION Low CSF ALLO levels in premenopausal women with PTSD might contribute to an imbalance in inhibitory versus excitatory neurotransmission, resulting in increased PTSD re-experiencing and depressive symptoms.
Collapse
Affiliation(s)
- Ann M Rasmusson
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | | | | | | | | | |
Collapse
|
422
|
Tsutsui K. Biosynthesis and organizing action of neurosteroids in the developing Purkinje cell. THE CEREBELLUM 2006; 5:89-96. [PMID: 16818383 DOI: 10.1080/14734220600697211] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Probing undiscovered neurosubstances that play important roles in the regulation of cerebellar function is essential for the progress of our understanding of the cerebellum. New findings over the past decade have established that the cerebellum as well as other brain regions synthesizes steroids de novo from cholesterol through mechanisms at least partly independent of peripheral steroidogenic glands. Such steroids synthesized de novo in the brain are called neurosteroids. Recently the Purkinje cell, a cerebellar neuron, has been identified as a major site for neurosteroid formation in the brain. This is the first demonstration of de novo neuronal neurosteroidogenesis in the brain. In mammals, the Purkinje cell actively synthesizes progesterone de novo from cholesterol during neonatal life, when cerebellar cortical formation occurs. 3alpha,5alpha-Tetrahydroprogesterone (allopregnanolone) is metabolized from progesterone in the neonatal cerebellum. Estrogen formation in the Purkinje cell may also occur in the neonate. Subsequently, recent studies on mammals using the Purkinje cell have demonstrated organizing actions of neurosteroids. Both progesterone and estradiol promote dendritic growth, spinogenesis and synaptogenesis via each cognate nuclear receptor in Purkinje neurons. Allopregnanolone is also involved in Purkinje and granule cell survival. Thus the Purkinje cell serves as an excellent cellular model for understanding the formation of cerebellar neuronal circuit in relation to organizing actions of neurosteroids.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Brain Science, Faculty of Integrated Arts and Sciences, Hiroshima University, and Integrative Brain Science Center at Hiroshima University, Higashi-Hiroshima, Japan.
| |
Collapse
|
423
|
Rasmusson AM, Picciotto MR, Krishnan-Sarin S. Smoking as a complex but critical covariate in neurobiological studies of posttraumatic stress disorders: a review. J Psychopharmacol 2006; 20:693-707. [PMID: 16401662 DOI: 10.1177/0269881106060193] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
As smoking rates in the general population continue to fall in response to new information and changing social values, the continued high rate of smoking among persons with psychiatric disorders has caught the attention of society at many levels: public health officials, medical and mental health care providers, and concerned family members alike. As a consequence, research studies aimed at quantifying the problem and understanding its cause have increased dramatically over the past several years. The following review first examines epidemiological studies that have revealed a bidirectional causal relationship between tobacco dependence and posttraumatic stress disorder (PTSD), one of several mental health disorders in which tobacco dependence remains prevalent and resistant to intervention. Second, we use a translational neuroscience perspective to discuss possible neurobiological mediators of the relationship between PTSD and tobacco dependence, hoping to spur further human and animal research that will elucidate pathogenetic mechanisms involved and inspire novel treatment interventions. Finally, to enable more effective clinical research in this area, we provide an overview of effective scientific methods for assessing and managing 'smoking status' as an experimental variable in clinical research studies of PTSD as well as other mental health disorders.
Collapse
Affiliation(s)
- Ann M Rasmusson
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| | | | | |
Collapse
|
424
|
Alexaki VI, Dermitzaki E, Charalampopoulos I, Kampa M, Nifli AP, Gravanis A, Margioris AN, Castanas E. Neuronal differentiation of PC12 cells abolishes the expression of membrane androgen receptors. Exp Cell Res 2006; 312:2745-56. [PMID: 16822503 DOI: 10.1016/j.yexcr.2006.04.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Revised: 04/01/2006] [Accepted: 04/06/2006] [Indexed: 11/29/2022]
Abstract
Sex steroids affect adrenal chromaffin cell function. In the present work, we have examined the expression and functional significance of membrane androgen receptor sites in normal rat adrenal chromaffin cells and in the PC12 rat pheochromocytoma cell line which can differentiate to either a neuronal or to an epithelial phenotype and expresses membrane estrogen receptor sites. Our data are as follows: (a) no cytosolic androgen receptors were found in both normal chromaffin and PC12 cells; (b) both types of chromaffin cells expressed high affinity membrane testosterone binding sites; (c) activation of these sites increased cytosolic Ca(2+), decreased catecholamine secretion and induced apoptosis; (d) NGF-induced neuronal differentiation of PC12 cells resulted in the suppression of the number of membrane testosterone sites. In conclusion, our data provide evidence for the existence of specific membrane testosterone receptors on adrenal chromaffin cells via which androgens, (some of them originating in the cortex) modulate their function. Neuronal differentiation of chromaffin cells results in a significant attenuation of these effects, via suppression of the expression of membrane androgen receptors suggesting, that the latter are specific for epithelioid chromaffin cells.
Collapse
Affiliation(s)
- Vassilia-Ismini Alexaki
- Laboratories of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion 71003, Greece
| | | | | | | | | | | | | | | |
Collapse
|
425
|
Yarim GF, Karahan S, Yarim M. Cerebellum progesterone concentration decreased in canine distemper virus infection. Res Vet Sci 2006; 82:173-80. [PMID: 16919304 DOI: 10.1016/j.rvsc.2006.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Revised: 04/27/2006] [Accepted: 05/20/2006] [Indexed: 11/26/2022]
Abstract
Progesterone has neuroprotective effects including augmentation of myelination in the central and peripheral nervous system. This study was designed to determine if demyelinating lesions in the cerebellum resulting from canine distemper virus (CDV) infection are associated with progesterone levels. Progesterone was measured using radioimmunoassay in samples of the cerebellum, corpus callosum, medulla oblongata, parietal, frontal, temporal, and occipital cortices as well as cerebrospinal fluid (CSF) and plasma collected from ten CDV infected and six non-infected dogs. The cerebellum progesterone level was significantly different between CDV infected (0.66+/-0.09 ng/g) and control dogs (1.14+/-0.09 ng/g) (p<0.001); however, no difference was observed for the other CNS regions, plasma and CSF (p>0.05). The cerebellum progesterone level was also significantly different between acute (0.71+/-0.0 5 ng/g) and chronic cases (0.61+/-0.09 ng/g) (p<0.05). The CDV infected cerebella were also categorized histopathologically according to the severity of demyelinating lesions as mild (n=5), moderate (n=2), or severe (n=3) among which the cerebellum progesterone level was significantly different (p<0.05). Progesterone concentration was 0.71+/-0.05 ng/g in mild, 0.65+/-0.10 ng/g in moderate, and 0.56+/-0.07 ng/g in severe cases. In conclusion, progesterone concentration decreases in the cerebellum in CDV infection and the severity of demyelinating lesions is the greatest in cerebella with the lowest progesterone concentrations. The results suggest that local impairment of progesterone metabolism may be associated with the initiation and progression of cerebellar lesions in CDV infection.
Collapse
Affiliation(s)
- Gul Fatma Yarim
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Ondokuz Mayis, 55139 Kurupelit, Samsun, Turkey.
| | | | | |
Collapse
|
426
|
Maurice T, Grégoire C, Espallergues J. Neuro(active)steroids actions at the neuromodulatory sigma1 (sigma1) receptor: biochemical and physiological evidences, consequences in neuroprotection. Pharmacol Biochem Behav 2006; 84:581-97. [PMID: 16945406 DOI: 10.1016/j.pbb.2006.07.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 06/30/2006] [Accepted: 07/07/2006] [Indexed: 01/05/2023]
Abstract
Steroids from peripheral sources or synthesized in the brain, i.e. neurosteroids, exert rapid modulations of neurotransmitter responses through specific interactions with membrane receptors, mainly the gamma-aminobutyric acid type A (GABA(A)) receptor and N-methyl-d-aspartate (NMDA) type of glutamate receptor. Progesterone and 3alpha-hydroxy-5alpha-pregnan-20-one (allopregnanolone) act as inhibitory steroids while pregnenolone sulfate or dehydroepiandrosterone sulfate act as excitatory steroids. Some steroids also interact with an atypical protein, the sigma(1) (sigma(1)) receptor. This receptor has been cloned in several species and is centrally expressed in neurons and oligodendrocytes. Activation of the sigma(1) receptor modulates cellular Ca(2+) mobilization, particularly from endoplasmic reticulum pools, and contributes to the formation of lipid droplets, translocating towards the plasma membrane and contributing to the recomposition of lipid microdomains. The present review details the evidences showing that the sigma(1) receptor is a target for neurosteroids in physiological conditions. Analysis of the sigma(1) protein sequence confirmed homologies with the ERG2/emopamil binding protein family but also with the steroidogenic enzymes isopentenyl diphosphate isomerase and 17beta-estradiol dehydrogenase. Biochemical and physiological arguments for an interaction of neuro(active)steroids with the sigma(1) receptor are analyzed and the impact on physiopathological outcomes in neuroprotection is illustrated.
Collapse
Affiliation(s)
- Tangui Maurice
- INSERM U. 710, Montpellier, F-34095 France University of Montpellier II, Montpellier, F-34095 France c EPHE, Paris, F-75007 France.
| | | | | |
Collapse
|
427
|
Meyer zu Hörste G, Prukop T, Nave KA, Sereda MW. Myelin disorders: Causes and perspectives of Charcot-Marie-Tooth neuropathy. J Mol Neurosci 2006; 28:77-88. [PMID: 16632877 DOI: 10.1385/jmn:28:1:77] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2005] [Revised: 11/30/1999] [Accepted: 08/08/2005] [Indexed: 01/25/2023]
Abstract
Charcot-Marie-Tooth (CMT) disease is a common hereditary neuropathy that causes progressive distally pronounced muscle weakness and can lead to life-long disability in patients. In most cases, the disorder has been associated with a partial duplication of human chromosome 17 (CMT1A), causing 1.5-fold overexpression of the peripheral myelin protein 22 kDa (PMP22). Increased PMP22 gene dosage results in demyelination, secondary axonal loss, and neurogenic muscle atrophy. Experimental therapeutic approaches based on the role of progesterone and ascorbic acid in myelin formation recently have reached preclinical proof-of-principle trials in rodents. It was shown that progesterone receptor antagonists can reduce PMP22 overexpression and clinical severity in a CMT1A rat model. Furthermore, ascorbic acid treatment reduced premature death and demyelination in a CMT1A mouse model. Thus, basic research has opened up new vistas for the understanding and treatment of hereditary neuropathies.
Collapse
|
428
|
Schlichter R, Keller AF, De Roo M, Breton JD, Inquimbert P, Poisbeau P. Fast nongenomic effects of steroids on synaptic transmission and role of endogenous neurosteroids in spinal pain pathways. J Mol Neurosci 2006; 28:33-51. [PMID: 16632874 DOI: 10.1385/jmn:28:1:33] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2005] [Revised: 11/30/1999] [Accepted: 06/28/2005] [Indexed: 11/11/2022]
Abstract
Steroids exert long-term modulatory effects on numerous physiological functions by acting at intracellular/nuclear receptors influencing gene transcription. Steroids and neurosteroids can also rapidly modulate membrane excitability and synaptic transmission by interacting with ion channels, that is, ionotropic neurotransmitter receptors or voltage-dependent Ca2+ or K+ channels. More recently, the cloning of a plasma membrane-located G protein-coupled receptor for progestins in various species has suggested that steroids/neurosteroids could also influence second-messenger pathways by directly interacting with specific membrane receptors. Here we review the experimental evidence implicating steroids/neurosteroids in the modulation of synaptic transmission and the evidence for a role of endogenously produced neurosteroids in such modulatory effects. We present some of our recent results concerning inhibitory synaptic transmission in lamina II of the spinal cord and show that endogenous 5alpha-reduced neurosteroids are produced locally in lamina II and modulate synaptic gamma-aminobutyric acid A(GABAA) receptor function during development, as well as during inflammatory pain. The production of 5alpha-reduced neurosteroids is controlled by the endogenous activation of the peripheral benzodiazepine receptor (PBR), which initiates the first step of neurosteroidogenesis by stimulating the translocation of cholesterol across the inner mitochondrial membrane. Tonic neurosteroidogenesis observed in immature animals was decreased during postnatal development, resulting in an acceleration of GABAA receptor-mediated miniature inhibitory postsynaptic current (mIPSC) kinetics observed in the adult. Stimulation of the PBR resulted in a prolongation of GABAergic mIPSCs at all ages and was observed during inflammatory pain. Neurosteroidogenesis might play an important role in the control of nociception at least at the spinal cord level.
Collapse
Affiliation(s)
- Rémy Schlichter
- Institut des Neurosciences Cellulaires et Intégratives-Centre National de la Recherche Scientifique (CNRS), Université Louis Pasteur, 67084 Strasbourg Cedex, France.
| | | | | | | | | | | |
Collapse
|
429
|
Do-Rego JL, Acharjee S, Seong JY, Galas L, Alexandre D, Bizet P, Burlet A, Kwon HB, Luu-The V, Pelletier G, Vaudry H. Vasotocin and mesotocin stimulate the biosynthesis of neurosteroids in the frog brain. J Neurosci 2006; 26:6749-60. [PMID: 16793882 PMCID: PMC6673836 DOI: 10.1523/jneurosci.4469-05.2006] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Revised: 04/25/2006] [Accepted: 04/25/2006] [Indexed: 11/21/2022] Open
Abstract
The neurohypophysial nonapeptides vasopressin (VP) and oxytocin (OT) modulate a broad range of cognitive and social activities. Notably, in amphibians, vasotocin (VT), the ortholog of mammalian VP, plays a crucial role in the control of sexual behaviors. Because several neurosteroids also regulate reproduction-related behaviors, we investigated the possible effect of VT and the OT ortholog mesotocin (MT) in the control of neurosteroid production. Double immunohistochemical labeling of frog brain sections revealed the presence of VT/MT-positive fibers in close proximity of neurons expressing the steroidogenic enzymes 3beta-hydroxysteroid dehydrogenase/delta5-delta4 isomerase (3beta-HSD) and cytochrome P450 17alpha-hydroxylase/c17, 20-lyase (P450(C17)). High concentrations of VT and MT receptor mRNAs were observed in diencephalic nuclei containing the 3beta-HSD and P450(C17) neuronal populations. Exposure of frog hypothalamic explants to graded concentrations of VT or MT produced a dose-dependent increase in the formation of progesterone, 17-hydroxypregnenolone, 17-hydroxyprogesterone, and dehydroepiandrosterone. The stimulatory effect of VT and MT on neurosteroid biosynthesis was mimicked by VP and OT, as well as by a selective V1b receptor agonist, whereas V2 and OT receptor agonists had no effect. VT-induced neurosteroid production was completely suppressed by selective V1a receptor antagonists and was not affected by V2 and OT receptor antagonists. Concurrently, the effect of MT on neurosteroidogenesis was markedly attenuated by selective OT and V1a receptor antagonists but not by a V2 antagonist. The present study provides the first evidence for a regulatory effect of VT and MT on neurosteroid biosynthesis. These data suggest that neurosteroids may mediate some of the behavioral actions of VT and MT.
Collapse
|
430
|
Citraro R, Russo E, Di Paola ED, Ibbadu GF, Gratteri S, Marra R, De Sarro G. Effects of some neurosteroids injected into some brain areas of WAG/Rij rats, an animal model of generalized absence epilepsy. Neuropharmacology 2006; 50:1059-71. [PMID: 16631210 DOI: 10.1016/j.neuropharm.2006.02.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2005] [Revised: 01/30/2006] [Accepted: 02/20/2006] [Indexed: 10/24/2022]
Abstract
Neurosteroids are synthesized in the brain and have been demonstrated to modulate various cerebral functions. Allopregnanolone (3alpha-hydroxy-5alpha-pregnan-20-one), a naturally occurring neurosteroid, and ganaxolone (3alpha-hydroxy-3beta-methyl-5alpha-pregnan-20-one), a synthetic derivative, are two neurosteroids acting as positive allosteric modulators of the GABA(A) receptor complex acting on a specific steroid recognition site. Both agents antagonize generalized tonic-clonic seizures in various animal models of epilepsy. Pregnenolone sulphate (3beta-hydroxy-5alpha-pregnen-20-one 3-sulphate; PS) is a negative allosteric modulator of GABA(A) receptors and a positive modulator of the NMDA receptors. We have evaluated the effects of such compounds in a genetic animal model of absence epilepsy, the WAG/Rij rat. Animals were chronically implanted with five frontoparietal cortical electrodes for electrocorticogram (EEG) recordings and bilateral guide cannulae into specific brain areas of the cortico-thalamic circuit in order to evaluate the effects of these compounds on the number and duration of epileptic spike-wave discharges (SWDs). The focal and bilateral microinjection of the two GABA(A) positive modulators into some thalamic nuclei (nucleus ventralis posteromedialis, nucleus reticularis thalami, nucleus ventralis posterolateralis was usually able to significantly worsen the occurrence of SWDs in WAG/Rij rats. Whereas both compounds were able to reduce the number and duration of SWDs when microinjected into the peri-oral region of the primary somatosensory cortex. The effects of PS were more complex depending on both the dose and the site of administration, generally, at low doses in thalamic nuclei and cortex, PS induced an increase of absence activity and a reduction at higher doses. These findings suggest that neurosteroids might play a role in absence epilepsies and that it might depend on the involvement of specific neuronal areas.
Collapse
Affiliation(s)
- Rita Citraro
- Section of Pharmacology, Department of Experimental and Clinical Medicine, Faculty of Medicine and Surgery, University of Catanzaro, Policlinico Mater Domini, Via T. Campanella, 115, 88100 Catanzaro, Italy
| | | | | | | | | | | | | |
Collapse
|
431
|
Eser D, Schüle C, Romeo E, Baghai TC, di Michele F, Pasini A, Zwanzger P, Padberg F, Rupprecht R. Neuropsychopharmacological properties of neuroactive steroids in depression and anxiety disorders. Psychopharmacology (Berl) 2006; 186:373-87. [PMID: 16247651 DOI: 10.1007/s00213-005-0188-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2005] [Accepted: 08/27/2005] [Indexed: 02/01/2023]
Abstract
Neuroactive steroids modulate neurotransmission through modulation of specific neurotransmitter receptors such as gamma-aminobutyric acid type A (GABAA) receptors. Preclinical studies suggested that neuroactive steroids may modulate anxiety- and depression-related behaviour and may contribute to the therapeutical effects of antidepressant drugs. Attenuations of 3alpha-reduced neuroactive steroids have been observed during major depression. This disequilibrium can be corrected by successful treatment with antidepressant drugs. However, non-pharmacological antidepressant treatment strategies did not affect neuroactive steroid composition independently from the clinical response. Further research is needed to clarify whether enhancement of neuroactive steroid levels might represent a new therapeutical approach in the treatment of affective disorders. Nevertheless, the first studies investigating the therapeutical effects of exogenously administered dehydroepiandosterone revealed promising results in the treatment of major depression. In addition, in various anxiety disorders alterations of neuroactive steroid levels have been observed. In panic disorder, in the absence of panic attacks, neuroactive steroid composition is opposite to that seen in depression, which may represent counter-regulatory mechanisms against the occurrence of spontaneous panic attacks. However, during experimentally induced panic attacks, there was a pronounced decline in GABAergic neuroactive steroids, which might contribute to the pathophysiology of panic attacks. In conclusion, neuroactive steroids contribute to the pathophysiology of affective disorders and the mechanisms of action of antidepressants. They are important endogenous modulators of depression and anxiety and may provide a basis for the development of novel therapeutic agents in the treatment of affective disorders.
Collapse
Affiliation(s)
- Daniela Eser
- Department of Psychiatry, Ludwig Maximilian University, Nussbaumstr. 7, 80336, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
432
|
Rasmusson AM, Wu R, Paliwal P, Anderson GM, Krishnan-Sarin S. A decrease in the plasma DHEA to cortisol ratio during smoking abstinence may predict relapse: a preliminary study. Psychopharmacology (Berl) 2006; 186:473-80. [PMID: 16609903 DOI: 10.1007/s00213-006-0367-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Accepted: 03/01/2006] [Indexed: 10/24/2022]
Abstract
RATIONALE Increases in depressive symptoms during smoking cessation have been associated with risk for relapse. Several studies have linked plasma levels of cortisol and dehydroepiandrosterone (DHEA) or DHEA-sulfate (DHEAS) to depressive symptoms. OBJECTIVES To determine whether changes in plasma cortisol, DHEA, or DHEAS levels and emergence of depressive symptoms during smoking cessation are associated with smoking relapse. MATERIALS AND METHODS Subjects were healthy non-medicated men and women, aged 39+/-12 years, who smoked, on average, 22 cigarettes per day. Depressive symptoms, smoking withdrawal symptoms, and plasma steroid levels were measured before and after 8 days of verified smoking abstinence. Relapse status at day 15 was then determined. RESULTS In the full sample (n=63), there was a trend for changes in depressive symptoms to be associated with relapse. In the subset of 25 subjects with plasma neuroactive steroid data, there was a significant interaction between the change in the plasma DHEA/cortisol ratio from day 0 to day 8 and relapse status at day 15. This ratio was similar before abstinence, but lower at day 8 in relapsed, compared to abstinent, subjects. Changes in the DHEA/cortisol ratio tended to predict changes in depressive symptoms in the women only. CONCLUSION A decrease in the plasma DHEA/cortisol ratio during 8 days of smoking abstinence was associated with relapse over the following week. Further research is needed to fully characterize sex-specific relationships between abstinence-induced changes in neuroactive steroid levels, depressive or withdrawal symptoms, and relapse. Such research may lead to new interventions for refractory smoking dependence.
Collapse
Affiliation(s)
- Ann M Rasmusson
- Department of Psychiatry, Yale University School of Medicine, New Haven, and Clinical Neuroscience Division, VA National Center for PTSD, West Haven, CT 06516, USA.
| | | | | | | | | |
Collapse
|
433
|
Mares P, Mikulecká A, Haugvicová R, Kasal A. Anticonvulsant action of allopregnanolone in immature rats. Epilepsy Res 2006; 70:110-7. [PMID: 16644184 DOI: 10.1016/j.eplepsyres.2006.03.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Revised: 03/14/2006] [Accepted: 03/18/2006] [Indexed: 10/24/2022]
Abstract
Anticonvulsant activity of allopregnanolone, a neurosteroid allosterically modulating GABA(A) receptor was tested in a model of motor seizures elicited by pentetrazol in immature rats. Rats 7, 12, 18, 25 or 90 days old were pretreated with allopregnanolone in doses from 5 to 40 mg/kg i.p. and 15 min later pentetrazol was injected subcutaneously in a dose of 100 mg/kg. Rats were observed in isolation for 30 min. Allopregnanolone dose-dependently suppressed both generalized tonic-clonic and minimal clonic seizures with the highest efficacy in 12-day-old rats. Anticonvulsant action was least expressed in adult animals. Duration of anticonvulsant action tested after a dose of 20 mg/kg in 12- and 90-day-old rats demonstrated markedly longer effects in young rats. Allopregnanolone compromised motor performance of rats but duration of this unwanted effect in 12-day-old rats was shorter than duration of anticonvulsant action. This difference can be important for possible clinical use of neurosteroids.
Collapse
Affiliation(s)
- P Mares
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | | | |
Collapse
|
434
|
Mo Q, Lu SF, Simon NG. Dehydroepiandrosterone and its metabolites: differential effects on androgen receptor trafficking and transcriptional activity. J Steroid Biochem Mol Biol 2006; 99:50-8. [PMID: 16524719 DOI: 10.1016/j.jsbmb.2005.11.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Accepted: 11/14/2005] [Indexed: 12/20/2022]
Abstract
Dehydroepiandrosterone (DHEA) is a multi-functional steroid that has been implicated in a broad range of biological effects in humans and rodents. Recent studies demonstrated that DHEA acts genomically through the androgen receptor (AR) in addition to its well-known effects on cell surface receptors. However, the relative contribution of DHEA and its major metabolites, including DHEA-Sulfate (DHEA-S), 7alpha-OH-DHEA, 7beta-OH-DHEA, 7-oxo-DHEA, androstenedione (Adione), and androstenediol (Adiol), in the production of genomic effects remains controversial, in part because the metabolism of DHEA varies in different cells and tissues. In the current study, the ability of DHEA and its metabolites to promote AR intracellular trafficking and regulate AR-mediated reporter gene expression, which are characteristic effects of androgens, was determined. Intracellular trafficking of AR-GFP protein was assessed in COS-7 cells while AR transcriptional activity was tested in CV-1 cells transiently co-transfected with AR expression plasmid and an MMTV-ARE-CAT reporter. The results demonstrated that DHEA, the 3beta-HSD metabolite Adione, and the 17beta-HSD metabolite Adiol, were androgenic. Each promoted AR-GFP intracellular trafficking, the formation of nuclear clusters, and AR-dependent transcriptional activity in a dose-dependent manner. In contrast, DHEA-S, 7alpha-OH-DHEA, 7beta-OH-DHEA, and 7-oxo-DHEA were ineffective and exhibited minimal androgenic activity, even at relatively high concentrations (10(-6) M). These results provide the first systematic comparison of the (i) androgenic activity of DHEA and its sulfated and hydroxylated metabolites, (ii) relative androgenicity of DHEA itself vs. the established androgens Adione and Adiol, and (iii) ability of DHEA and its major metabolites to promote AR-GFP intracellular trafficking. In addition to partitioning DHEA and its metabolites into compounds with (DHEA, Adione, Adiol) and without (DHEA-S, 7alpha-OH-DHEA, 7beta-OH-DHEA, and 7-oxo-DHEA) androgenic activity, the findings improve our understanding of the intracellular processes mediating the genomic effects of DHEA through AR.
Collapse
Affiliation(s)
- Qianxing Mo
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015, United States
| | | | | |
Collapse
|
435
|
King SR, Smith AGA, Alpy F, Tomasetto C, Ginsberg SD, Lamb DJ. Characterization of the putative cholesterol transport protein metastatic lymph node 64 in the brain. Neuroscience 2006; 139:1031-8. [PMID: 16549269 DOI: 10.1016/j.neuroscience.2006.01.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Revised: 12/20/2005] [Accepted: 01/15/2006] [Indexed: 11/25/2022]
Abstract
Intracellular management of cholesterol is a critical process in the brain. Deficits with cholesterol transport and storage are linked to neurodegenerative disorders such as Neimann-Pick disease type C and Alzheimer's disease. One protein putatively involved in cholesterol transport is metastatic lymph node 64 (MLN64). MLN64 localizes to late endosomes which are part of the cholesterol internalization pathway. However, a detailed pattern of MLN64 expression in the brain is unclear. Using immunocytochemical and immunoblot analyses, we demonstrated the presence of MLN64 in several tissue types and various regions within the brain. MLN64 immunostaining in the CNS was heterogeneous, indicating selective expression in discrete specific cell populations and regions. MLN64 immunoreactivity was detected in glia and neurons, which displayed intracellular labeling consistent with an endosomal localization. Although previous studies suggested that MLN64 may promote steroid production in the brain, MLN64 immunoreactivity did not colocalize with steroidogenic cells in the CNS. These results demonstrate that MLN64 is produced in the mouse and human CNS in a restricted pattern of expression, suggesting that MLN64 serves a cell-specific function in cholesterol transport.
Collapse
Affiliation(s)
- S R King
- Scott Department of Urology, Room N730, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
436
|
Lavaque E, Sierra A, Azcoitia I, Garcia-Segura LM. Steroidogenic acute regulatory protein in the brain. Neuroscience 2006; 138:741-7. [PMID: 16338087 DOI: 10.1016/j.neuroscience.2005.05.060] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Accepted: 05/31/2005] [Indexed: 01/19/2023]
Abstract
The nervous system synthesizes steroids that regulate the development and function of neurons and glia, and have neuroprotective properties. The first step in steroidogenesis involves the delivery of free cholesterol to the inner mitochondrial membrane where it can be converted into pregnenolone by the enzyme cytochrome P450side chain cleavage. The peripheral-type benzodiazepine receptor and the steroidogenic acute regulatory protein are involved in this process and appear to function in a coordinated manner. Steroidogenic acute regulatory protein mRNA and protein are widely expressed throughout the adult brain. Steroidogenic acute regulatory protein expression has been detected in many neuronal populations, in ependymocytes, in some astroglial cells, in Schwann cells from peripheral nerves and in proliferating cells of the developing and adult brain. Steroidogenic acute regulatory protein is colocalized in the same neural cells with P450side chain cleavage and with other steroidogenic enzymes. Steroidogenic acute regulatory protein expression in the brain shows marked changes with development, aging and injury. The steroidogenic acute regulatory protein gene may be under the control of diverse mechanisms in different neural cell types, since its expression is upregulated by cyclic AMP (cAMP) in gliomas and astrocytes in culture and downregulated by cyclic AMP (cAMP) in Schwann cells. In addition, activation of N-methyl-D-aspartate receptors, and the consequent rise in intracellular calcium levels, activates steroidogenic acute regulatory protein and steroidogenesis in hippocampal neurons. In conclusion, steroidogenic acute regulatory protein is regulated in the nervous system by different physiological and pathological conditions and may play an important role during brain development, aging and after injury.
Collapse
Affiliation(s)
- E Lavaque
- Instituto Cajal, CSIC, Madrid, Spain
| | | | | | | |
Collapse
|
437
|
Debatin T, Barbosa ADE. Effect of isopregnanolone on rapid tolerance to the anxiolytic effect of ethanol. BRAZILIAN JOURNAL OF PSYCHIATRY 2006; 28:18-23. [PMID: 16612485 DOI: 10.1590/s1516-44462006000100005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJETIVE: It has been shown that neurosteroids can either block or stimulate the development of chronic and rapid tolerance to the incoordination and hypothermia caused by ethanol consumption. The aim of the present study was to investigate the influence of isopregnanolone on the development of rapid tolerance to the anxiolytic effect of ethanol in mice. METHOD: Male Swiss mice were pretreated with isopregnanolone (0.05, 0.10 or 0.20 mg/kg) 30 min before administration of ethanol (1.5 g/kg). Twenty-four hours later, all animals we tested using the plus-maze apparatus. The first experiment defined the doses of ethanol that did or did not induce rapid tolerance to the anxiolytic effect of ethanol. In the second, the influence of pretreatment of mice with isopregnanolone (0.05, 0.10 or 0.20 mg/kg) on rapid tolerance to ethanol (1.5 g/kg) was studied. CONCLUSIONS: The results show that pretreatment with isopregnanolone interfered with the development of rapid tolerance to the anxiolytic effect of ethanol.
Collapse
Affiliation(s)
- Thaize Debatin
- Department of Natural Sciences, Universidade Regional de Blumenau, Blumenau, SC, Brazil
| | | |
Collapse
|
438
|
Lim G, Wang S, Zeng Q, Sung B, Yang L, Mao J. Expression of spinal NMDA receptor and PKCgamma after chronic morphine is regulated by spinal glucocorticoid receptor. J Neurosci 2006; 25:11145-54. [PMID: 16319314 PMCID: PMC6725649 DOI: 10.1523/jneurosci.3768-05.2005] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Spinal NMDA receptor (NMDAR), protein kinase C (PKC), and glucocorticoid receptor (GR) have all been implicated in the mechanisms of morphine tolerance; however, how these cellular elements interact after chronic morphine exposure remains unclear. Here we show that the expression of spinal NMDAR and PKCgamma after chronic morphine is regulated by spinal GR through a cAMP response element-binding protein (CREB)-dependent pathway. Chronic morphine (10 microg, i.t.; twice daily for 6 d) induced a time-dependent upregulation of GR, the NR1 subunit of NMDAR, and PKCgamma within the rat's spinal cord dorsal horn. This NR1 and PKCgamma upregulation was significantly diminished by intrathecal coadministration of morphine with the GR antagonist RU38486 or a GR antisense oligodeoxynucleotide. Intrathecal coadministration of morphine with an adenylyl cyclase inhibitor (2',5'-dideoxyadenosine) or a protein kinase A inhibitor (H89) also significantly attenuated morphine-induced NR1 and PKCgamma expression, whereas intrathecal treatment with an adenylyl cyclase activator (forskolin) alone mimicked morphine-induced expression of GR, NR1, and PKCgamma. Moreover, the expression of phosphorylated CREB was upregulated within the spinal cord dorsal horn after chronic morphine, and a CREB antisense oligodeoxynucleotide coadministered intrathecally with morphine prevented the upregulation of GR, NR1, and PKCgamma. These results indicate that spinal GR through the cAMP-CREB pathway played a significant role in NMDAR and PKCgamma expression after chronic morphine exposure. The data suggest that genomic interaction among spinal GR, NMDAR, and PKCgamma may be an important mechanism that contributes to the development of morphine tolerance.
Collapse
Affiliation(s)
- Grewo Lim
- Pain Research Group, Division of Pain Medicine, Department of Anesthesia and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | |
Collapse
|
439
|
Xie C, Richardson JA, Turley SD, Dietschy JM. Cholesterol substrate pools and steroid hormone levels are normal in the face of mutational inactivation of NPC1 protein. J Lipid Res 2006; 47:953-63. [PMID: 16461760 DOI: 10.1194/jlr.m500534-jlr200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mutational inactivation of NPC1 largely blocks the movement of LDL-derived cholesterol from the lysosome to the metabolically active, cytosolic pool of sterol that is the substrate for steroid hormone production. Such a block might, in theory, lead to deficiencies in circulating levels of testosterone, progesterone, and corticosterone. However, there are at least two other sources for cellular cholesterol, de novo synthesis and scavenger receptor class B type I-mediated uptake of HDL cholesteryl ester (CE). In this study, we measured the rates of net cholesterol acquisition by these three pathways in the adrenal, ovary, and testis. In all three organs, the majority (81-98%) of cholesterol acquisition came from the selective uptake of CE from HDL and de novo synthesis. Furthermore, in the npc1(-/-)mouse, the cytosolic storage pool of CE in a tissue such as the adrenal remained constant (approximately 25 mg/g). As a result of these alternative pathways, the plasma concentrations of testosterone (3.5 vs. 2.5 ng/ml), progesterone (8.5 vs. 6.7 ng/ml), and corticosterone (391 vs. 134 ng/ml) were either the same or elevated in the npc1(-/-)mouse, compared with the control animal. Thus, impairment of cholesterol acquisition through the NPC1-dependent, clathrin-coated pit pathway did not limit the availability of cholesterol substrate for steroid hormone synthesis in the steroidogenic cells.
Collapse
Affiliation(s)
- Chonglun Xie
- Department of Internal Medicine, University of Texas Southwestern Medical School, Dallas, 75390-8887, USA
| | | | | | | |
Collapse
|
440
|
Johansson T, Frändberg PA, Nyberg F, Le Grevès P. Low concentrations of neuroactive steroids alter kinetics of [3H]ifenprodil binding to the NMDA receptor in rat frontal cortex. Br J Pharmacol 2006; 146:894-902. [PMID: 16170326 PMCID: PMC1751215 DOI: 10.1038/sj.bjp.0706397] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The modulatory effects of the two neurosteroids pregnenolone sulphate (PS) and pregnanolone sulphate (3alpha5betaS) on [3H]ifenprodil binding to the N-methyl-D-aspartate (NMDA) receptor in rat frontal cortex were studied. The binding for [3H]ifenprodil itself displayed monophasic kinetics in all experiments. None of the neurosteroids displaced the radioligand from its binding site on the NR2B subunit of the NMDA receptor. However, their continual presence at nanomolar concentrations had significant effects on ligand binding kinetics, interacting through distinct sites in saturation, competition and dissociation experiments. PS at 30 nM enhanced the specific binding to about 150% of that in its absence and enhanced the dissociation rate three-fold indicating a positive modulation of [3H]ifenprodil binding to the NMDA receptor. Furthermore, PS increased Bmax and decreased Kd suggesting that the neurosteroid exposes new [3H]ifenprodil binding sites with altered properties. In contrast, 3alpha5betaS (30 nM) decreased specific [3H]ifenprodil binding to approximately 40% of that determined for the radioligand alone. The presence of 3alpha5betaS at nanomolar concentrations induced biphasic curve fits in saturation, competition as well as dissociation experiments. In conclusion, the present study show that the allosteric modulators PS or 3alpha5betaS change [3H]ifenprodil binding kinetics in a way indicating conformational alteration of its binding site on the NR2B subunit.
Collapse
Affiliation(s)
- Tobias Johansson
- Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, P.O. Box 591, S-751 24 Uppsala, Sweden
| | | | | | | |
Collapse
|
441
|
Ritsner M, Gibel A, Ram E, Maayan R, Weizman A. Alterations in DHEA metabolism in schizophrenia: two-month case-control study. Eur Neuropsychopharmacol 2006; 16:137-46. [PMID: 16139994 DOI: 10.1016/j.euroneuro.2005.07.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2005] [Accepted: 07/21/2005] [Indexed: 11/29/2022]
Abstract
OBJECTIVE The goals of this study were to determine whether alterations in serum dehydroepiandrosterone (DHEA), its sulfated conjugate (DHEAS), androstenedione, testosterone, and progesterone concentrations occur in schizophrenia patients compared with healthy controls over two months, and their associations with psychopathology, emotional distress, and antipsychotic treatment. METHOD Serum hormones were repeatedly determined for 21 antipsychotic-treated male DSM-IV schizophrenia patients and 14 healthy controls. Observations were at four time points: upon entry into the study, and after 2, 4 and 8 weeks. RESULTS In schizophrenia patients compared with healthy controls serum concentration of DHEA and androstenedione found increased, but that of DHEAS decreased, while progesterone and testosterone showed normal levels. Schizophrenia patients were also characterized by elevated androstenedione/DHEAS molar ratios, and reduced DHEAS/DHEA and testosterone/androstenedione molar ratios compared with healthy controls. Concentrations and molar ratios of serum hormones did not significantly change during the study either among schizophrenia patients, or healthy controls. Among patients alterations in DHEA, DHEAS and androstenedione were associated with emotional distress, anxiety, dysphoric mood, positive and activation symptoms, serum prolactin levels, but were not related to age, antipsychotic agents, and extrapyramidal side effects. CONCLUSIONS Alterations in DHEA metabolism in schizophrenia are attributed to the distress, anxiety, severity of symptoms, prolactin levels, and may represent a marker for impaired hormonal responses to stress. These findings should be considered when evaluating the discrepancies in DHEA studies in schizophrenia.
Collapse
Affiliation(s)
- Michael Ritsner
- Acute Department & Clinical Psychobiology Laboratory, Sha'ar Menashe Mental Health Center, Mobile Post Hefer, Hadera, Israel.
| | | | | | | | | |
Collapse
|
442
|
Prange-Kiel J. Östrogenwirkungen im Gehirn. GYNAKOLOGISCHE ENDOKRINOLOGIE 2006. [DOI: 10.1007/s10304-005-0132-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
443
|
Huang MH, So EC, Liu YC, Wu SN. Glucocorticoids stimulate the activity of large-conductance Ca2+ -activated K+ channels in pituitary GH3 and AtT-20 cells via a non-genomic mechanism. Steroids 2006; 71:129-40. [PMID: 16274717 DOI: 10.1016/j.steroids.2005.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Revised: 08/23/2005] [Accepted: 09/07/2005] [Indexed: 12/18/2022]
Abstract
The effects of glucocorticoids on ion currents were investigated in pituitary GH3 and AtT-20 cells. In whole-cell configuration, dexamethasone, a synthetic glucocorticoid, reversibly increased the density of Ca2+ -activated K+ current (IK(Ca)) with an EC50 value of 21 +/- 5 microM. Dexamethasone-induced increase in IK(Ca) density was suppressed by paxilline (1 microM), yet not by glibenclamide (10 microM), pandinotoxin-Kalpha (1 microM) or mifepristone (10 microM). Paxilline is a blocker of large-conductance Ca2+ -activated K+ (BKCa) channels, while glibenclamide and pandinotoxin-Kalpha are blockers of ATP-sensitive and A-type K+ channels, respectively. Mifepristone can block cytosolic glucocorticoid receptors. In inside-out configuration, the application of dexamethasone (30 microM) into the intracellular surface caused no change in single-channel conductance; however, it did increase BKCa -channel activity. Its effect was associated with a negative shift of the activation curve. However, no Ca2+ -sensitiviy of these channels was altered by dexamethasone. Dexamethasone-stimulated channel activity involves an increase in mean open time and a decrease in mean closed time. Under current-clamp configuration, dexamethasone decreased the firing frequency of action potentials. In pituitary AtT-20 cells, dexamethasone (30 microM) also increased BKCa -channel activity. Dexamethasone-mediated stimulation of IK(Ca) presented here that is likely pharmacological, seems to be not linked to a genomic mechanism. The non-genomic, channel-stimulating properties of dexamethasone may partly contribute to the underlying mechanisms by which glucocorticoids affect neuroendocrine function.
Collapse
Affiliation(s)
- Mei-Han Huang
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, No. 1, University Road, Tainan, Taiwan
| | | | | | | |
Collapse
|
444
|
Charalampopoulos I, Alexaki VI, Lazaridis I, Dermitzaki E, Avlonitis N, Tsatsanis C, Calogeropoulou T, Margioris AN, Castanas E, Gravanis A. G protein-associated, specific membrane binding sites mediate the neuroprotective effect of dehydroepiandrosterone. FASEB J 2006; 20:577-9. [PMID: 16407456 DOI: 10.1096/fj.05-5078fje] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The neurosteroid dehydroepiandrosterone (DHEA) at 1 nM protects NMDA-/GABAA-receptor negative neural crest-derived PC12 cells from apoptosis. We now report that membrane-impermeable DHEA-BSA conjugate replaces unconjugated DHEA in protecting serum-deprived PC12 cells from apoptosis (IC50=1.5 nM). Protection involves phosphorylation of the prosurvival factor Src and induction of the anti-apoptotic protein Bcl-2 and is sensitive to pertussis toxin. Binding assays of [3H]DHEA on isolated PC12 cell membranes revealed saturation within 30 min and binding of DHEA with a Kd of 0.9 nM. A similar binding activity was detectable in isolated membranes from rat hippocampus and from normal human adrenal chromaffin cells. The presence of DHEA-specific membrane binding sites was confirmed by flow cytometry and confocal laser microscopy of DHEA-BSA-FITC stained cells. In contrast to estrogens and progestins, glucocorticoids and androgens displaced DHEA from its membrane binding sites but with a 10-fold lower affinity than DHEA (IC50=9.3 and 13.6 nM, respectively). These agents acted as pure antagonists, blocking the antiapoptotic effect of DHEA as well as the induction of Bcl-2 proteins and Src kinase activation. In conclusion, our findings suggest that neural crest-derived cells possess specific DHEA membrane binding sites coupled to G proteins. Binding to these sites confers neuroprotection.
Collapse
|
445
|
Verleye M, Akwa Y, Liere P, Ladurelle N, Pianos A, Eychenne B, Schumacher M, Gillardin JM. The anxiolytic etifoxine activates the peripheral benzodiazepine receptor and increases the neurosteroid levels in rat brain. Pharmacol Biochem Behav 2006; 82:712-20. [PMID: 16388839 DOI: 10.1016/j.pbb.2005.11.013] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 11/10/2005] [Accepted: 11/24/2005] [Indexed: 11/23/2022]
Abstract
The peripheral benzodiazepine receptors (PBR) might be involved in certain pathophysiological events, such as anxiety, by stimulating the production of neuroactive steroids in the brain. A recent electrophysiological study has revealed an interaction between PK11195, a PBR ligand and the anxiolytic compound etifoxine at micromolar concentrations. The present work was aimed at further characterizing the etifoxine-PBR interaction. In membrane preparations from intact male rat forebrain, etifoxine uncompetitively inhibited the binding of [(3)H]PK11195 with an IC(50) = 18.3 +/- 1.2 microM, a value consistent with etifoxine plasma and brain concentrations measured after an anxiolytic-like dose (50 mg/kg). In vivo, that etifoxine dose was associated with increased concentrations of pregnenolone, progesterone, 5alpha-dihydroprogesterone and allopregnanolone in plasma and brain of sham-operated animals. In adrenalectomized and castrated rats, etifoxine enhanced the brain levels of these steroids, suggesting a stimulation of their local synthesis and/or a decrease of their disappearance rate, independently of peripheral sources. Finasteride, an inhibitor of 5alpha-reductase that converts progesterone into its 5alpha-reduced metabolites like allopregnanolone, attenuated the anti-conflict effect of etifoxine even though brain allopregnanolone contents were drastically reduced. These results indicate that following activation of the PBR in the brain, an increased cerebral production of allopregnanolone, a potent positive modulator of the GABA(A) receptor function, may partially contribute to the anxiolytic-like effects of etifoxine.
Collapse
Affiliation(s)
- Marc Verleye
- Département de Pharmacologie, Biocodex, Zac de Mercières, Chemin d'Armancourt, Compiègne, France.
| | | | | | | | | | | | | | | |
Collapse
|
446
|
Simon NG, Mo Q, Hu S, Garippa C, Lu SF. Hormonal Pathways Regulating Intermale And Interfemale Aggression. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2006; 73:99-123. [PMID: 16737902 DOI: 10.1016/s0074-7742(06)73003-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Affiliation(s)
- Neal G Simon
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | | | | | | | | |
Collapse
|
447
|
Monnet FP, Maurice T. The Sigma1 Protein as a Target for the Non-genomic Effects of Neuro(active)steroids: Molecular, Physiological, and Behavioral Aspects. J Pharmacol Sci 2006; 100:93-118. [PMID: 16474209 DOI: 10.1254/jphs.cr0050032] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Steroids synthesized in the periphery or de novo in the brain, so called 'neurosteroids', exert both genomic and nongenomic actions on neurotransmission systems. Through rapid modulatory effects on neurotransmitter receptors, they influence inhibitory and excitatory neurotransmission. In particular, progesterone derivatives like 3alpha-hydroxy-5alpha-pregnan-20-one (allopregnanolone) are positive allosteric modulators of the gamma-aminobutyric acid type A (GABA(A)) receptor and therefore act as inhibitory steroids, while pregnenolone sulphate (PREGS) and dehydroepiandrosterone sulphate (DHEAS) are negative modulators of the GABA(A) receptor and positive modulators of the N-methyl-D-aspartate (NMDA) receptor, therefore acting as excitatory neurosteroids. Some steroids also interact with atypical proteins, the sigma (sigma) receptors. Recent studies particularly demonstrated that the sigma1 receptor contributes effectively to their pharmacological actions. The present article will review the data demonstrating that the sigma1 receptor binds neurosteroids in physiological conditions. The physiological relevance of this interaction will be analyzed and the impact on physiopathological outcomes in memory and drug addiction will be illustrated. We will particularly highlight, first, the importance of the sigma1-receptor activation by PREGS and DHEAS which may contribute to their modulatory effect on calcium homeostasis and, second, the importance of the steroid tonus in the pharmacological development of selective sigma1 drugs.
Collapse
Affiliation(s)
- François P Monnet
- Unité 705 de l'Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 7157 du Centre National de la Recherche Scientifique, Université de Paris V et VII, Hôpital Lariboisière-Fernand Widal, Paris, France
| | | |
Collapse
|
448
|
Ebner MJ, Corol DI, Havlíková H, Honour JW, Fry JP. Identification of neuroactive steroids and their precursors and metabolites in adult male rat brain. Endocrinology 2006; 147:179-90. [PMID: 16223859 DOI: 10.1210/en.2005-1065] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Steroids in the brain arise both from local synthesis and from peripheral sources and have a variety of effects on neuronal function. However, there is little direct chemical evidence for the range of steroids present in brain or of the pathways for their synthesis and inactivation. This information is a prerequisite for understanding the regulation and function of brain steroids. After extraction from adult male rat brain, we have fractionated free steroids and their sulfate esters and then converted them to heptafluorobutyrate or methyloxime-trimethylsilyl ether derivatives for unequivocal identification and assay by gas chromatography analysis and selected ion monitoring mass spectrometry. In the free steroid fraction, corticosterone, 3alpha,5alpha-tetrahydrodeoxycorticosterone, testosterone, and dehydroepiandrosterone were found in the absence of detectable precursors usually found in endocrine glands, indicating peripheral sources and/or alternative synthetic pathways in brain. Conversely, the potent neuroactive steroid 3alpha,5alpha-tetrahydroprogesterone (allopregnanolone) was found in the presence of its precursors pregnenolone, progesterone, and 5alpha-dihydroprogesterone. Furthermore, the presence of 3beta-, 11beta-, 17alpha-, and 20alpha-hydroxylated metabolites of 3alpha,5alpha-tetrahydroprogesterone implicated possible inactivation pathways for this steroid. The 20alpha-reduced metabolites could also be found for pregnenolone, progesterone, and 5alpha-dihydroprogesterone, introducing a possible regulatory diversion from the production of 3alpha,5alpha-tetrahydroprogesterone. In the steroid sulfate fraction, dehydroepiandrostrone sulfate was identified but not pregnenolone sulfate. Although pharmacologically active, identification of the latter appears to be an earlier methodological artifact, and the compound is thus of doubtful physiological significance in the adult brain. Our results provide a basis for elucidating the origins and regulation of brain steroids.
Collapse
Affiliation(s)
- M J Ebner
- Department of Physiology, University College London, London WC1E 6BT, United Kingdom
| | | | | | | | | |
Collapse
|
449
|
Tsutsui K, Matsunaga M, Miyabara H, Ukena K. Neurosteroid biosynthesis in the quail brain: a review. ACTA ACUST UNITED AC 2006; 305:733-42. [PMID: 16902960 DOI: 10.1002/jez.a.302] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The brain traditionally has been considered to be a target site of peripheral steroid hormones. In contrast to this classical concept, new findings over the past decade have shown that the brain itself also has the capability of forming steroids de novo, the so-called "neurosteroids". De novo neurosteroidogenesis in the brain from cholesterol is a conserved property of vertebrates. Our studies using the quail, as an excellent animal model, have demonstrated that the avian brain possesses cytochrome P450 side-chain cleavage enzyme (P450scc), 3beta-hydroxysteroid dehydrogenase/Delta(5)-Delta(4)-isomerase (3beta-HSD), cytochrome P450 17alpha-hydroxylase/c17,20-lyase (P450(17alpha,lyase)), 17beta-HSD, etc., and produces pregnenolone, progesterone, 3beta, 5beta-tetrahydroprogesterone, androstenedione, testosterone and estradiol from cholesterol. However, the biosynthetic pathway of neurosteroids in the avian brain from cholesterol may be still incomplete, because we recently found that the quail brain actively produces 7alpha-hydroxypregnenolone, a previously undescribed avian neurosteroid. This paper summarize the advances made in our understanding of biosynthesis of neurosteroids in the avian brain.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Brain Science, Faculty of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan.
| | | | | | | |
Collapse
|
450
|
Birzniece V, Bäckström T, Johansson IM, Lindblad C, Lundgren P, Löfgren M, Olsson T, Ragagnin G, Taube M, Turkmen S, Wahlström G, Wang MD, Wihlbäck AC, Zhu D. Neuroactive steroid effects on cognitive functions with a focus on the serotonin and GABA systems. ACTA ACUST UNITED AC 2005; 51:212-39. [PMID: 16368148 DOI: 10.1016/j.brainresrev.2005.11.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Revised: 11/10/2005] [Accepted: 11/11/2005] [Indexed: 01/20/2023]
Abstract
This article will review neuroactive steroid effects on serotonin and GABA systems, along with the subsequent effects on cognitive functions. Neurosteroids (such as estrogen, progesterone, and allopregnanolone) are synthesized in the central and peripheral nervous system, in addition to other tissues. They are involved in the regulation of mood and memory, in premenstrual syndrome, and mood changes related to hormone replacement therapy, as well as postnatal and major depression, anxiety disorders, and Alzheimer's disease. Estrogen and progesterone have their respective hormone receptors, whereas allopregnanolone acts via the GABA(A) receptor. The action of estrogen and progesterone can be direct genomic, indirect genomic, or non-genomic, also influencing several neurotransmitter systems, such as the serotonin and GABA systems. Estrogen alone, or in combination with antidepressant drugs affecting the serotonin system, has been related to improved mood and well being. In contrast, progesterone can have negative effects on mood and memory. Estrogen alone, or in combination with progesterone, affects the brain serotonin system differently in different parts of the brain, which can at least partly explain the opposite effects on mood of those hormones. Many of the progesterone effects in the brain are mediated by its metabolite allopregnanolone. Allopregnanolone, by changing GABA(A) receptor expression or sensitivity, is involved in premenstrual mood changes; and it also induces cognitive deficits, such as spatial-learning impairment. We have shown that the 3beta-hydroxypregnane steroid UC1011 can inhibit allopregnanolone-induced learning impairment and chloride uptake potentiation in vitro and in vivo. It would be important to find a substance that antagonizes allopregnanolone-induced adverse effects.
Collapse
Affiliation(s)
- Vita Birzniece
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University Hospital, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|