Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Neurotherapeutics ; 9(3): 635-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22454323

ABSTRACT

Recent work exploring the use of high-molecular weight alcohols to treat essential tremor (ET) has identified octanoic acid as a potential novel tremor-suppressing agent. We used an established harmaline-based mouse model of ET to compare tremor suppression by 1-octanol and octanoic acid. The dose-related effect on digitized motion power within the tremor bandwidth as a fraction of overall motion power was analyzed. Both 1-octanol and octanoic acid provided significant reductions in harmaline tremor. An 8-carbon alkyl alcohol and carboxylic acid each suppress tremor in a pre-clinical mouse model of ET. Further studies are warranted to determine the safety and efficacy of such agents in humans with ET.


Subject(s)
Caprylates/therapeutic use , Central Nervous System Stimulants/toxicity , Essential Tremor/chemically induced , Essential Tremor/drug therapy , Harmaline/toxicity , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred ICR
2.
J Neurosci Res ; 89(3): 394-405, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21259326

ABSTRACT

Recently, a variant of insulin-like growth factor-1, mechano-growth factor (MGF), has been discovered whose 24-amino-acid carboxy end is protective in models of stroke, nerve injury, and amyotrophic lateral sclerosis, suggesting broad-spectrum neuroprotective properties. Moreover, we recently demonstrated in vitro and in vivo that a modified protease-resistant 24-amino-acid MGF derivative (MGF24) protects dopaminergic neurons from oxidative stress-induced apoptosis via induction of the stress response protein heme oxygenase-1. However, the underlying mechanism by which MGF24 up-regulates heme oxygenase-1 expression is unknown. In this study, we demonstrate that MGF24-induced heme oxygenase-1 up-regulation is dependent on activation of protein kinase Cϵ and NF-E2-related factor-2 (Nrf2). MGF24 induces nuclear translocation of Nrf2, and siRNA knockdown of Nrf2 or of heme oxygenase-1 prevents MGF24-induced heme oxygenase-1 up-regulation and neuroprotection of SH-SY5Y cells against 6-hydroxydopamine-induced cell death. Pharmacological inhibition of ERK, p38 MAPK, PI3K/Akt, or PKC signaling revealed that only PKC inhibition by GF109203X prevents MGF24's ability to protect against 6-hydroxydopamine-induced cell death. GF109203X also prevented MGF24-induced Nrf2 nuclear translocation and heme oxygenase-1 up-regulation. siRNA knockdown of protein kinase Cϵ blocks MGF24-induced Nfr2 nuclear translocation, heme oxygenase-1 expression, and neuroprotection. Taken together, these results demonstrate that PKC activity is needed for MGF24's activation of Nrf2, which in turn increases heme oxygenase-1 expression, a critical event in mediating MGF24's neuroprotection against 6-hydroxydopamine-induced apoptosis.


Subject(s)
Heme Oxygenase-1/metabolism , Insulin-Like Growth Factor I/metabolism , NF-E2-Related Factor 2/metabolism , Protein Kinases/metabolism , Up-Regulation/physiology , Analysis of Variance , Cell Line, Tumor , Cell Survival/drug effects , Heme Oxygenase-1/genetics , Humans , Insulin-Like Growth Factor I/chemistry , NF-E2-Related Factor 2/genetics , Neuroblastoma , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , Peptide Fragments/pharmacology , Protein Kinases/genetics , Protein Transport/drug effects , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Sympatholytics/toxicity , Transfection/methods , Up-Regulation/drug effects
3.
Eur J Pharmacol ; 659(1): 30-6, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21256842

ABSTRACT

NNC 55-0396 [(1S,2S)-2-(2-(N-[(3-benzimidazol-2-yl)propyl]-N-methylamino)ethyl)-6-fluoro-1,2, 3,4-tetrahydro-1-isopropyl-2-naphtyl cyclopropanecarboxylate dihydrochloride], is a mibefradil derivative that retains potent in vitro T-type calcium channel antagonist efficacy. We compared the two compounds for behavioral toxicity, effects on cytochrome P450 activity, and efficacy against tremor in the γ-aminobutyric acid type A (GABAA) receptor subunit α1-null mouse, and the harmaline tremor model of essential tremor in wild-type mice. NNC 55-0396 was better tolerated than mibefradil in the horizontal wire test of sedation/motor function, with 3/6 failing at 300 and 30mg/kg respectively. To assess for a potential interaction with harmaline, mice were given the drugs, followed by harmaline or vehicle, and tested 30min later in the inverted wire grid test. Mibefradil exacerbated, whereas NNC 55-0396 ameliorated harmaline-induced test deficits. In mouse liver microsomes, NNC 55-0396 was a less potent inhibitor of harmaline O-demethylation than mibefradil (Ki: 0.95 and 0.29µM respectively), and also less potent at inhibiting testosterone 6-ß-hydroxylation (Ki: 0.71 and 0.12µM respectively). In the GABAA α1-null model, NNC 55-0396 but not mibefradil, (each at 20mg/kg), suppressed tremor while NNC 55-0396 at 12.5mg/kg suppressed harmaline-induced tremor by half by 20-100min, whereas mibefradil at the same dose did not significantly affect tremor. In contrast to mibefradil, NNC 55-0396 is well tolerated and suppresses tremor, and exerts less cytochrome P450 inhibition. These results suggest potential clinical utility for NNC 55-0396 or similar derivatives as a T-type calcium antagonist.


Subject(s)
Behavior, Animal/drug effects , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Essential Tremor/drug therapy , Mibefradil/chemistry , Mibefradil/pharmacology , Naphthalenes/chemistry , Naphthalenes/pharmacology , Animals , Benzimidazoles/therapeutic use , Cyclopropanes/therapeutic use , Disease Models, Animal , Essential Tremor/enzymology , Essential Tremor/metabolism , Gene Deletion , Harmaline/metabolism , Hydroxylation/drug effects , Methylation/drug effects , Mibefradil/therapeutic use , Mice , Naphthalenes/therapeutic use , Receptors, GABA-A/deficiency , Receptors, GABA-A/genetics , Structure-Activity Relationship , Testosterone/metabolism
4.
Clin Neuropharmacol ; 33(5): 223-6, 2010.
Article in English | MEDLINE | ID: mdl-20838216

ABSTRACT

OBJECTIVE: We studied the potential efficacy and tolerance of memantine for essential tremor in an open-treatment trial. METHODS: Participants with upper-limb tremor were titrated to no more than 40 mg/d memantine, as monotherapy or as adjunct to stable antitremor medication, followed by a 12-week extension phase. Tremor was assessed in study 1 with accelerometry and in study 2 by blinded ratings of videotaped Washington Heights Inwood Genetic Essential Tremor (WHIGET) rating scale items. Subjects also rated their tremor treatment response and tremor-associated impairment on the Functional Disabilities scale. RESULTS: In study 1, average accelerometry-measured tremor at last titration visit (average dose, 30.3 mg/d) did not change from baseline, but 2 of 9 subjects, taking 40 mg/d, had greater than 70% accelerometry tremor reduction. In study 2, 13 of 16 provided evaluable data. Average blinded rater-evaluated WHIGET scores were significantly different from baseline scores among those taking 20 mg/d (-12.7%; P < 0.05), but not at last titration visit (-8.4%; average dose, 30.4 mg/d), 40 mg/d (-14.1%), or at end-of-extension visit (-18.2%). Raters judged WHIGET scores as greater than 30% improved in 2 subjects. Unblinded subjects rated Functional Disabilities significantly improved at 30 to 40 but not at 10 to 20 mg/d, and tremor treatment response was positive at all doses. Adverse events were more common at higher doses and included dizziness, somnolence, and poor energy. CONCLUSIONS: These pilot results with small samples indicate that the average effect of memantine on tremor is mild or not significant. However, in a small subset of patients, memantine may confer meaningful tremor benefit.


Subject(s)
Dopamine Agents/therapeutic use , Essential Tremor/drug therapy , Memantine/therapeutic use , Aged , Disability Evaluation , Drug Therapy, Combination , Essential Tremor/physiopathology , Female , Humans , Male , Pilot Projects , Severity of Illness Index , Treatment Outcome , Upper Extremity/physiopathology
5.
Neuropharmacology ; 59(6): 380-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20547167

ABSTRACT

Essential tremor is a common disorder that lacks molecular targets for therapeutic development. T-type calcium channel activation has been postulated to underlie rhythmicity in the olivo-cerebellar system that is implicated in essential tremor. We therefore tested whether compounds that antagonize T-type calcium channel currents suppress tremor in two mouse models that possess an essential tremor-like pharmacological response profile. Tremor was measured using digitized spectral motion power analysis with harmaline-induced tremor and in the GABA(A) receptor α1 subunit-null model. Mice were given ethosuximide, zonisamide, the neuroactive steroid (3ß,5α,17ß)-17-hydroxyestrane-3-carbonitrile (ECN), the 3,4-dihydroquinazoline derivative KYS05064, the mibefradil derivative NNC 55-0396, or vehicle. In non-sedating doses, each compound reduced harmaline-induced tremor by at least 50% (range of maximal suppression: 53-81%), and in the GABA(A) α1-null model by at least 70% (range 70-93%). Because the T-type calcium channel Cav3.1 is the dominant subtype expressed in the inferior olive, we assessed the tremor response of Cav3.1-deficient mice to harmaline, and found that null and heterozygote mice exhibit as much tremor as wild-type mice. In addition, ECN and NNC 55-0396 suppressed harmaline tremor as well in Cav3.1-null mice as in wild-type mice. The finding that five T-type calcium antagonists suppress tremor in two animal tremor models suggests that T-type calcium channels may be an appropriate target for essential tremor therapy development. It is uncertain whether medications developed to block only the Cav3.1 subtype would exhibit efficacy.


Subject(s)
Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/metabolism , Essential Tremor/drug therapy , Analysis of Variance , Animals , Benzimidazoles/therapeutic use , Cyclopropanes/therapeutic use , Disease Models, Animal , Essential Tremor/chemically induced , Essential Tremor/metabolism , Estranes/therapeutic use , Ethosuximide/therapeutic use , Harmaline , Isoxazoles/therapeutic use , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Naphthalenes/therapeutic use , Nitriles/therapeutic use , Receptors, GABA-A/metabolism , Zonisamide
6.
Exp Neurol ; 220(2): 255-66, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19735655

ABSTRACT

To assess potential efficacy of mechano growth factor (MGF) for chronic neurodegenerative disorders, we studied whether MGF protects dopamine (DA) neurons subjected to neurotoxic stress. We show that a short 24-amino acid C-terminal peptide of MGF (MGF24) upregulates heme oxygenase-1 (HO-1) expression and protects SH-SY5Y cells against apoptosis and cell loss induced by three DA cell-specific neurotoxins: 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenylpyridinium (MPP(+)), and rotenone. MGF24 maintains the mitochondrial membrane potential and blocks the release of mitochondrial apoptotic-inducing factor into the cytoplasm induced by 6-OHDA, MPP(+), and rotenone. Chemical inhibition of HO-1 with zinc protoporphyrin-IX prevents neuroprotection by MGF24 against the three neurotoxins. MGF24 does not activate Akt signaling nor does Akt inhibition block MGF24 protection of SH-SY5Y cells. In 6-OHDA-lesioned rats, central or peripheral MGF24 administration protects against the development of contralateral forelimb under-utilization, reduces ipsilateral nigral DA cell body loss, and attenuates tyrosine hydroxylase fiber loss in the ipsilateral striatum, independent of IGF-1 receptor activation. Peripheral MGF24 administration upregulates HO-1 expression in striatal and midbrain tissue. This report is the first to demonstrate that a small peptide, MGF24, upregulates HO-1, an important cell defense mediator, and protects DA cells, suggesting new strategies for neuroprotection in Parkinson's disease.


Subject(s)
Dopamine/physiology , Heme Oxygenase-1/biosynthesis , Neurons/drug effects , Neuroprotective Agents , STAT5 Transcription Factor/pharmacology , Tumor Suppressor Proteins/pharmacology , Animals , Cell Line , DNA Fragmentation/drug effects , Enzyme Induction/drug effects , Insulin-Like Growth Factor I/metabolism , Membrane Potentials/drug effects , Mitochondrial Membranes/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Neurons/enzymology , Oxidopamine/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Sympatholytics/toxicity , Tetrazolium Salts , Thiazoles , Up-Regulation/drug effects
7.
Drug Metab Dispos ; 36(7): 1291-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18411403

ABSTRACT

A novel mibefradil derivative, NNC55-0396, designed to be hydrolysis-resistant, was shown to be a selective T-type Ca(2+) channel inhibitor without L-type Ca(2+) channel efficacy. However, its effects on cytochromes P450 (P450s) have not previously been examined. We investigated the inhibitory effects of NNC55-0396 toward seven major recombinant human P450s--CYP3A4, CYP2D6, CYP1A2, CYP2C9, CYP2C8, CYPC19, and CYP2E1--and compared its effects with those of mibefradil and its hydrolyzed metabolite, Ro40-5966. Our results show that CYP3A4 and CYP2D6 are the two P450s most affected by mibefradil, Ro40-5966, and NNC55-0396. Mibefradil (IC(50) = 33 +/- 3 nM, K(i) = 23 +/- 0.5 nM) and Ro40-5966 (IC(50) = 30 +/- 7.8 nM, K(i) = 21 +/- 2.8 nM) have a 9- to 10-fold greater inhibitory activity toward recombinant CYP3A4 benzyloxy-4-trifluoromethylcoumarin-O-debenzylation activity than NNC55-0396 (IC(50) = 300 +/- 30 nM, K(i) = 210 +/- 6 nM). More dramatically, mibefradil (IC(50) = 566 +/- 71 nM, K(i) = 202 +/- 39 nM) shows 19-fold higher inhibition of CYP3A-associated testosterone 6beta-hydroxylase activity in human liver microsomes compared with NNC55-0396 (IC(50) = 11 +/- 1.1 microM, K(i) = 3.9 +/- 0.4 microM). Loss of testosterone 6beta-hydroxylase activity by recombinant CYP3A4 was shown to be time- and concentration-dependent with both compounds. However, NNC55-0396 (K(I) = 3.87 microM, K(inact) = 0.061/min) is a much less potent mechanism-based inhibitor than mibefradil (K(I) = 83 nM, K(inact) = 0.048/min). In contrast, NNC55-0396 (IC(50) = 29 +/- 1.2 nM, K(i) = 2.8 +/- 0.3 nM) and Ro40-5966 (IC(50) = 46 +/- 11 nM, K(i) = 4.5 +/- 0.02 nM) have a 3- to 4-fold greater inhibitory activity toward recombinant CYP2D6 than mibefradil (IC(50) = 129 +/- 21 nM, K(i) = 12.7 +/- 0.9 nM). Our results suggest that NNC55-0396 could be a more favorable T-type Ca(2+) antagonist than its parent compound, mibefradil, which was withdrawn from the market because of strong inhibition of CYP3A4.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Cytochrome P-450 CYP3A Inhibitors , Mibefradil/chemistry , Cytochrome P-450 CYP3A , Humans , Hydrolysis , Mibefradil/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Recombinant Proteins/antagonists & inhibitors
8.
Dev Neurobiol ; 68(5): 632-44, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18278798

ABSTRACT

Recently, using the medial forebrain bundle (MFB) 6-hydroxydopmaine (6-OHDA) lesion rat model of Parkinson's disease (PD), we have demonstrated that blockade of central IGF-1 receptors (IGF-1R) attenuated estrogen neuroprotection of substantia nigra pars compacta (SNpc) DA neurons, but exacerbated 6-OHDA lesions in IGF-1 only treated rats (Quesada and Micevych [2004]: J Neurosci Res 75:107-116). This suggested that the IGF-1 system is a central mechanism through which estrogen acts to protect the nigrostriatal DA system. Moreover, these results also suggest that IGF-1R-induced intracellular signaling pathways are involved in the estrogen mechanism that promotes neuronal survival. In vitro, two convergent intracellular signaling pathways used by estrogen and IGF-1, the mitogen-activated protein kinase (MAPK/ERK), and phosphatidyl-inositol-3-kinase/Akt (PI3K/Akt), have been demonstrated to be neuroprotective. Continuous central infusions of MAPK/ERK and PI3K/Akt inhibitors were used to test the hypothesis that one or both of these signal transduction pathways mediates estrogen and/or IGF-1 neuroprotection of SNpc DA neurons after a unilateral administration of 6-OHDA into the MFB of rats. Motor behavior tests and tyrosine hydroxylase immunoreactivity revealed that the inhibitor of the PI3K/Akt pathway (LY294002) blocked the survival effects of both estrogen and IGF-1, while an inhibitor of the MAPK/ERK signaling (PD98059) was ineffective. Western blot analyses showed that estrogen and IGF-1 treatments increased PI3K/Akt activation in the SN; however, MAPK/ERK activation was decreased in the SN. Indeed, continuous infusions of inhibitors blocked phosphorylation of PI3K/Akt and MAPK/ERK. These findings indicate that estrogen and IGF-1-mediated SNpc DA neuronal protection is dependent on PI3K/Akt signaling, but not on the MAPK/ERK pathway.


Subject(s)
Dopamine/physiology , Estrogens/physiology , Insulin-Like Growth Factor I/physiology , Neurons/physiology , Oncogene Protein v-akt/metabolism , Parkinson Disease, Secondary/pathology , Phosphatidylinositol 3-Kinases/metabolism , Substantia Nigra/physiology , Animals , Blotting, Western , Cell Count , Enzyme Activation/drug effects , Estradiol/pharmacology , Female , Injections, Intraventricular , Motor Activity/drug effects , Motor Activity/physiology , Nerve Fibers/enzymology , Ovariectomy , Parkinson Disease, Secondary/chemically induced , Rats , Rats, Long-Evans , Signal Transduction/drug effects , Signal Transduction/physiology , Substantia Nigra/cytology , Tyrosine 3-Monooxygenase/metabolism
9.
Neuroendocrinology ; 88(1): 35-42, 2008.
Article in English | MEDLINE | ID: mdl-18212517

ABSTRACT

Sex steroids modulate reproduction by altering the response of steroid-activated opioid circuits in the hypothalamus and limbic system, by inducing release of endogenous opioids and activation of their cognate receptors. Many studies have concentrated on steroid regulation of exogenous opioid peptides, but steroids also have important actions on opioid receptors inducing receptor trafficking. Opioid receptors are G protein-coupled receptors and their activation catalyzes the exchange of GTP for GDP initiating intracellular signaling cascades. Kinetics of G protein activation were studied using [(35)S]GTPgammaS binding. Catalytic amplification, the number of G proteins activated per occupied receptor, was used as a measure of receptor/transducer amplification. The present study examined whether estrogen and progesterone treatment altered the kinetics of nociceptin opioid receptor (ORL1) in plasma membranes from the medial preoptic area and mediobasal hypothalamus. These hypothalamic regions are important in the gonadal steroid hormone regulation of sexual receptivity. In the mediobasal hypothalamus, estrogen increased ORL1 (B(max)) receptor number 2-fold and maximal GTPgammaS binding (E(max)) 3.9-fold. Subsequent progesterone treatment further increased ORL1 E(max )6.9-fold above baseline, despite a 2-fold decrease in the catalytic amplification factor. In the medial preoptic area, estrogen alone did not increase E(max), but both estrogen and progesterone were able to increase ORL1 B(max) 2.2-fold and E(max) 3-fold, despite having a 3-fold decrease in the catalytic amplification factor. These effects are interesting because they indicate actions of steroids that increase the number of ORL1 but decrease the catalytic amplification suggesting that the steroid effects on opioid receptors are complex and may involve modulation by other signals.


Subject(s)
Estrogens/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Progesterone/physiology , Receptors, Opioid/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Estrogens/pharmacology , Female , Guanosine 5'-O-(3-Thiotriphosphate)/antagonists & inhibitors , Guanosine 5'-O-(3-Thiotriphosphate)/biosynthesis , Hypothalamus/drug effects , Hypothalamus/metabolism , Narcotic Antagonists , Opioid Peptides/metabolism , Opioid Peptides/physiology , Progesterone/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Long-Evans , Sulfur Radioisotopes/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , Nociceptin Receptor , Nociceptin
10.
J Comp Neurol ; 503(1): 198-208, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17480015

ABSTRACT

Although several studies have focused on the neuroprotective effects of estrogen (E2) on stroke, there have been tantalizing reports on the potential neuroprotective role of E2 in degenerative neuronal diseases such as Alzheimer's and Parkinson's (PD). In animal models of PD, E2 protects the nigrostriatal dopaminergic (DA) system against neurotoxins. However, little is known about the cellular and molecular mechanism(s) involved by which E2 elicits its neuroprotective effects on the nigrostriatal DA system. A preferred mechanism for neuroprotection is the interaction of E2 with specific neuroprotective growth factors and receptors. One such neuroprotective factor/receptor system is insulin-like growth factor-1 (IGF-1). E2 neuroprotective effects in the substantia nigra (SN) DA system have been shown to be dependent on IGF-1. To determine whether E2 also interacts with the IGF-1 receptor (IGF-1R) and to determine the cellular localization of estrogen receptor (ER) and IGF-1R, we compared the distribution of ER and IGF-1R in the SN. Stereological measurements revealed that 40% of the subpopulation of tyrosine hydroxylase-immunoreactive (TH-ir) SN pars compacta (SNpc) DA neurons are immunoreactive for estrogen receptor-beta (ERbeta). No immunolabeling for ERalpha was observed. In situ hybridization and immunocytochemistry studies confirmed the expression of IGF-1R mRNA and revealed that almost all TH-ir SNpc DA neurons were immunoreactive for IGF-1R, respectively. Moreover, one-third of glial fibrillary acidic protein (GFAP-ir) cells in the SN were ERbeta-ir, and 67% of GFAP-ir cells expressed IGF-1R-ir. Therefore, the localization of ERbeta and IGF-1R on SNpc DA neurons and astrocytes suggests a modulatory role of E2 on IGF-1R, and this modulation may affect neuroprotection.


Subject(s)
Estrogen Receptor beta/metabolism , Neuroglia/metabolism , Neurons/metabolism , Receptor, IGF Type 1/metabolism , Substantia Nigra/metabolism , Animals , Dopamine/metabolism , Female , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Rats , Rats, Long-Evans , Substantia Nigra/cytology , Tyrosine 3-Monooxygenase/metabolism
11.
Brain Res ; 1073-1074: 316-20, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16472782

ABSTRACT

Cholecystokinin (CCK) in the nervous system has effects opposite to those of opioids. However, the mechanism by which CCK opposes the effect of opioids at the receptor or cellular level is still unknown. In the brain, distributions of CCK receptors and opioid receptors have been demonstrated to overlap. The present study was undertaken to determine the mechanism of CCK-opioid interactions in the cortex of ovariectomized rats. Furthermore, because estrogen is a powerful regulator of CCK and opioid activity, we examined whether estrogen state also modulates the interactions of these neuropeptides. mu-Opioid (MOP) receptor binding was examined in cortical membranes that were preincubated with CCK-8S and CCK receptor agonist and antagonist followed with 3H-DAMGO. Pharmacological results revealed that CCK-8S suppressed 3H-DAMGO binding in cortical membranes of ovariectomized rats. The same result was obtained using a CCK1 receptor agonist (JMV-180), whereas a CCK2 receptor agonist (CCK-4) failed to suppress 3H-DAMGO binding. Antagonism of the CCK1 receptor by JMV-179 blocked both CCK-8S and JMV-180 suppression of 3H-DAMGO binding. Furthermore, estrogen treatment to female rats resulted in a suppression of 3H-DAMGO binding in cortical membranes. These results demonstrate an estrogen regulation of the MOP receptor and a protein-protein interaction between CCK1 receptor and MOP receptor.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Estrogens/pharmacology , Receptors, Cholecystokinin/physiology , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacokinetics , Analysis of Variance , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Interactions , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Female , Hormone Antagonists/pharmacology , Ovariectomy/methods , Proglumide/analogs & derivatives , Proglumide/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Radioligand Assay/methods , Rats , Rats, Long-Evans , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/antagonists & inhibitors , Sincalide/analogs & derivatives , Sincalide/pharmacology , Tritium/pharmacokinetics
12.
J Neurosci Res ; 75(1): 107-16, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14689453

ABSTRACT

The most prominent neurochemical hallmark of Parkinson's disease (PD) is the loss of nigrostriatal dopamine (DA). Animal models of PD have concentrated on depleting DA and therapies have focused on maintaining or restoring DA. Within this context estrogen protects against 6-hydroxdopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) lesions of the nigrostriatal DA pathway. Present studies tested the hypothesis that neuroprotective estrogen actions involve activation of the insulin-like growth factor-1 (IGF-1) system. Ovariectomized rats were treated with either a single subcutaneous injection of 17beta-estradiol benzoate or centrally or peripherally IGF-1. All rats were infused unilaterally with 6-OHDA into the medial forebrain bundle (MFB) to lesion the nigrostriatal DA pathway. Tyrosine hydroxylase (TH) immunocytochemistry confirmed that rats injected with 6-OHDA had a massive loss of TH immunoreactivity in both the ipsilateral substantia nigra compacta (60% loss) and the striatum (>95% loss) compared to the contralateral side. Loss of TH immunoreactivity was correlated with loss of asymmetric forelimb movements, a behavioral assay for motor deficits. Pretreatment with estrogen or IGF-1 significantly prevented 6-OHDA-induced loss of substantia nigra compacta neurons (20% loss) and TH immunoreactivity in DA fibers in the striatum (<20% loss) and prevented the loss of asymmetric forelimb use. Blockage of IGF-1 receptors by intracerebroventricular JB-1, an IGF-1 receptor antagonist, attenuated both estrogen and IGF-1 neuroprotection of nigrostriatal DA neurons and motor behavior. These findings suggest that IGF-1 and estrogen acting through the IGF-1 system may be critical for neuroprotective effects of estrogen on nigrostriatal DA neurons in this model of PD.


Subject(s)
Dopamine/metabolism , Estrogens/pharmacology , Insulin-Like Growth Factor I/pharmacology , Motor Activity/drug effects , Neostriatum/drug effects , Oxidopamine/toxicity , Animals , Cell Count , Drug Administration Routes , Drug Interactions , Estradiol/pharmacology , Female , Insulin-Like Growth Factor I/antagonists & inhibitors , Medial Forebrain Bundle/injuries , Neostriatum/metabolism , Ovariectomy , Rats , Rats, Long-Evans , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
13.
J Neurosci ; 22(6): 2401-8, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11896179

ABSTRACT

The ovarian hormone estradiol (E(2)) and insulin-like growth factor-I (IGF-I) interact in the CNS to regulate neuroendocrine function and synaptic remodeling. Previously, our laboratory showed that 2 d E(2) treatment induces alpha(1B)-adrenoceptor expression and promotes IGF-I enhancement of alpha(1)-adrenoceptor potentiation of cAMP accumulation in the preoptic area (POA) and hypothalamus (HYP). This study examined the hypothesis that E(2)-dependent aspects of female reproductive function, including alpha(1B)-adrenoceptor expression and function in the POA and HYP, are mediated by brain IGF-I receptors (IGF-IRs) in female rats. Ovariohysterectomized rats were implanted with a guide cannula aimed at the third ventricle and treated in vivo with vehicle or E(2) daily for 2 d before experimentation. Intracerebroventricular infusions of JB-1, a selective IGF-IR antagonist, were administered every 12 hr beginning 1 hr before the first E(2) injection. Administration of JB-1 during E(2) priming completely blocks hormone-induced luteinizing hormone release and partially inhibits hormone-dependent reproductive behavior. Reproductive behavior is restored by intracerebroventricular infusion of 8-bromo-cGMP, the second messenger implicated in alpha(1)-adrenergic facilitation of lordosis. In addition, blockade of IGF-IRs during E(2) priming prevents E(2)-induced increases in alpha(1B)-adenoceptor binding density and abolishes acute IGF-I enhancement of NE-stimulated cAMP accumulation in HYP and POA slices. These data document the existence of a novel mechanism by which IGF-I participates in the remodeling of noradrenergic receptor signaling in the HYP and POA after E(2) treatment. These events may help coordinate the timing of ovulation with the expression of sexual receptivity.


Subject(s)
Cyclic GMP/analogs & derivatives , Estradiol/metabolism , Insulin-Like Growth Factor I/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Reproduction/physiology , Adrenergic alpha-1 Receptor Antagonists , Animals , Binding, Competitive/drug effects , Cyclic GMP/pharmacology , Estradiol/administration & dosage , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Hysterectomy , In Vitro Techniques , Injections, Intraventricular , Insulin-Like Growth Factor I/pharmacology , Male , Norepinephrine/pharmacology , Ovariectomy , Peptides/administration & dosage , Posture , Prazosin/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/antagonists & inhibitors , Sexual Behavior, Animal/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
14.
Pharmacol Toxicol ; 91(6): 387-97, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12688384

ABSTRACT

Responses of the nervous system to introceptive and extroceptive inputs depend upon the state of the brain. Oestrogen has the ability to modulate brain state and dramatically alter interactions among neural circuits to influence an organism's responses to given stimuli. Cholecystokinin (CCK) and endogenous opioid peptides (EOP) have a wide and parallel distribution in the nervous system. Their reciprocal interactions regulate a diverse physiology including reproduction, cortical function and nociception. The actions of CCK and EOP are diametrically opposed, in many regions. For example, when opioids inhibit reproductive behaviour or nociception, CCK facilitates. Because oestrogen is a powerful regulator of the expression of CCK and EOP, we examined whether oestrogen-state also modulated the interactions of these neuropeptides. In this paper we present new data and review previous work that demonstrates oestrogen modulation of functional CCK-opioid interactions that regulate reproductive behaviour, cortical function and nociception.


Subject(s)
Brain , Central Nervous System , Cholecystokinin/physiology , Estrogens/physiology , Opioid Peptides/physiology , Animals , Brain/drug effects , Brain/metabolism , Central Nervous System/drug effects , Central Nervous System/physiology , Cholecystokinin/metabolism , Drug Interactions , Female , Lordosis/chemically induced , Opioid Peptides/metabolism , Rats , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...