151
|
Hannapel RC, Henderson YH, Nalloor R, Vazdarjanova A, Parent MB. Ventral hippocampal neurons inhibit postprandial energy intake. Hippocampus 2017; 27:274-284. [PMID: 28121049 DOI: 10.1002/hipo.22692] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2016] [Indexed: 12/12/2022]
Abstract
Evidence suggests that the memory of a recently ingested meal limits subsequent intake. Given that ventral hippocampal (vHC) neurons are involved in memory and energy intake, the present experiment tested the hypothesis that vHC neurons contribute to the formation of a memory of a meal and inhibit energy intake during the postprandial period. We tested (1) whether pharmacological inactivation of vHC neurons during the period following a sucrose meal, when the memory of the meal would be undergoing consolidation, accelerates the onset of the next sucrose meal and increases intake and (2) whether sucrose intake increases vHC expression of the synaptic plasticity marker activity-regulated cytoskeletal-associated protein (Arc). Adult male Sprague-Dawley rats were trained to consume a 32% sucrose solution daily at the same time and location. On the experimental day, the rats were given intra-vHC infusions of the GABAA receptor agonist muscimol or vehicle after they finished their first sucrose meal. Compared to vehicle infusions, postmeal intra-vHC muscimol infusions decreased the latency to the next sucrose meal, increased the amount of sucrose consumed during that meal, increased the total number of sucrose meals and the total amount of sucrose ingested. In addition, rats that consumed sucrose had higher levels of Arc expression in both vHC CA1 and CA3 subfields than cage control rats. Collectively, these findings are the first to show that vHC neurons inhibit energy intake during the postprandial period and support the hypothesis that vHC neurons form a memory of a meal and inhibit subsequent intake. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Yoko H Henderson
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| | - Rebecca Nalloor
- Neuroscience Institute, Augusta Biomedical Research Corporation, Charlie Norwood VA Medical Center, 950 15th Street, Augusta, Georgia
| | - Almira Vazdarjanova
- Department of Pharmacology and Toxicology, Augusta University, 1120 15th Street, CB 3526, Augusta, Georgia.,VA Research Service, Charlie Norwood VA Medical Center, 950 15th Street, Augusta, Georgia
| | - Marise B Parent
- Neuroscience Institute, Georgia State University, Atlanta, Georgia.,Department of Psychology, Georgia State University, Atlanta, Georgia
| |
Collapse
|
152
|
Pardo J, Abba MC, Lacunza E, Francelle L, Morel GR, Outeiro TF, Goya RG. Identification of a conserved gene signature associated with an exacerbated inflammatory environment in the hippocampus of aging rats. Hippocampus 2017; 27:435-449. [DOI: 10.1002/hipo.22703] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Joaquín Pardo
- INIBIOLP, Histology B–Pathology B, School of Medicine, UNLPLa Plata Argentina
| | | | | | - Laetitia Francelle
- Department of Neurodegeneration and Restorative ResearchUniversity Medical Center GöttingenGöttingen Germany
| | - Gustavo R. Morel
- INIBIOLP, Histology B–Pathology B, School of Medicine, UNLPLa Plata Argentina
| | - Tiago F. Outeiro
- Department of Neurodegeneration and Restorative ResearchUniversity Medical Center GöttingenGöttingen Germany
| | - Rodolfo G. Goya
- INIBIOLP, Histology B–Pathology B, School of Medicine, UNLPLa Plata Argentina
| |
Collapse
|
153
|
Zancan M, Dall'Oglio A, Quagliotto E, Rasia‐Filho AA. Castration alters the number and structure of dendritic spines in the male posterodorsal medial amygdala. Eur J Neurosci 2016; 45:572-580. [DOI: 10.1111/ejn.13460] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/22/2016] [Accepted: 10/31/2016] [Indexed: 01/21/2023]
Affiliation(s)
- Mariana Zancan
- Department of Basic Sciences/Physiology Federal University of Health Sciences Sarmento Leite 245 Porto Alegre RS 90050‐170 Brazil
- Graduation Program in Neuroscience Federal University of Rio Grande do Sul Porto Alegre Brazil
| | - Aline Dall'Oglio
- Department of Basic Sciences/Physiology Federal University of Health Sciences Sarmento Leite 245 Porto Alegre RS 90050‐170 Brazil
| | - Edson Quagliotto
- Department of Basic Sciences/Physiology Federal University of Health Sciences Sarmento Leite 245 Porto Alegre RS 90050‐170 Brazil
| | - Alberto A. Rasia‐Filho
- Department of Basic Sciences/Physiology Federal University of Health Sciences Sarmento Leite 245 Porto Alegre RS 90050‐170 Brazil
- Graduation Program in Neuroscience Federal University of Rio Grande do Sul Porto Alegre Brazil
| |
Collapse
|
154
|
Gurevich EV, Gainetdinov RR, Gurevich VV. G protein-coupled receptor kinases as regulators of dopamine receptor functions. Pharmacol Res 2016; 111:1-16. [PMID: 27178731 PMCID: PMC5079267 DOI: 10.1016/j.phrs.2016.05.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/03/2016] [Accepted: 05/06/2016] [Indexed: 02/08/2023]
Abstract
Actions of the neurotransmitter dopamine in the brain are mediated by dopamine receptors that belong to the superfamily of G protein-coupled receptors (GPCRs). Mammals have five dopamine receptor subtypes, D1 through D5. D1 and D5 couple to Gs/olf and activate adenylyl cyclase, whereas D2, D3, and D4 couple to Gi/o and inhibit it. Most GPCRs upon activation by an agonist are phosphorylated by GPCR kinases (GRKs). The GRK phosphorylation makes receptors high-affinity binding partners for arrestin proteins. Arrestin binding to active phosphorylated receptors stops further G protein activation and promotes receptor internalization, recycling or degradation, thereby regulating their signaling and trafficking. Four non- visual GRKs are expressed in striatal neurons. Here we describe known effects of individual GRKs on dopamine receptors in cell culture and in the two in vivo models of dopamine-mediated signaling: behavioral response to psychostimulants and L-DOPA- induced dyskinesia. Dyskinesia, associated with dopamine super-sensitivity of striatal neurons, is a debilitating side effect of L-DOPA therapy in Parkinson's disease. In vivo, GRK subtypes show greater receptor specificity than in vitro or in cultured cells. Overexpression, knockdown, and knockout of individual GRKs, particularly GRK2 and GRK6, have differential effects on signaling of dopamine receptor subtypes in the brain. Furthermore, deletion of GRK isoforms in select striatal neuronal types differentially affects psychostimulant-induced behaviors. In addition, anti-dyskinetic effect of GRK3 does not require its kinase activity: it is mediated by the binding of its RGS-like domain to Gαq/11, which suppresses Gq/11 signaling. The data demonstrate that the dopamine signaling in defined neuronal types in vivo is regulated by specific and finely orchestrated actions of GRK isoforms.
Collapse
Affiliation(s)
- Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37221, USA.
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia; Skolkovo Institute of Science and Technology, Skolkovo, 143025, Moscow, Russia
| | | |
Collapse
|
155
|
Vrecl M, Jorgensen R, Pogacnik A, Heding A. Development of a BRET2 Screening Assay Using β-Arrestin 2 Mutants. ACTA ACUST UNITED AC 2016; 9:322-33. [PMID: 15191649 DOI: 10.1177/1087057104263212] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study has focused on enhancing the signal generated from the interaction between a G-protein-coupled receptor (GPCR) and β-arrestin 2 (β-arr2), measured by the bioluminescence resonance energy transfer (BRET2) technology. Both class A (β2-adrenergic receptor [β2-AR]) and class B (neurokinin-type 1 receptor [NK1-R]) GPCRs, classified based on their internalization characteristics, have been analyzed. It was evaluated whether the BRET2 signal can be enhanced by using (1) β-arr2 phosphorylation-independent mutant (β-arr2 R169E) and (2) β-arr2 mutants deficient in their ability to interact with the components of the clathrin-coated vesicles (β-arr2 R393E, R395E and β-arr2 373 stop). For the class B receptor, there was no major difference in the agonist-promoted BRET2 signal when comparing results obtained with wild-type (wt) and mutant β-arr2. However, with the class A receptor, a more than 2-fold increase in the BRET2 signal was observed with β-arr2 mutants lacking the AP-2 or both AP-2 and clathrin binding sites. This set of data suggests that the inability of these β-arr2 mutants to interact with the components of the clathrin-coated vesicle probably prevents their rapid dissociation from the receptor, thus yielding an increased and more stable BRET2 signal. The β-arr2 R393E, R395E mutant also enhanced the signal window with other members of the GPCR family (neuropeptide Y type 2 receptor [NPY2-R] and TG1019 receptor) and was successfully applied in full-plate BRET2-based agonist and antagonist screening assays.
Collapse
Affiliation(s)
- Milka Vrecl
- Institute of Anatomy, Histology & Embryology, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | | | | |
Collapse
|
156
|
Agster KL, Tomás Pereira I, Saddoris MP, Burwell RD. Subcortical connections of the perirhinal, postrhinal, and entorhinal cortices of the rat. II. efferents. Hippocampus 2016; 26:1213-30. [PMID: 27101786 DOI: 10.1002/hipo.22600] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2016] [Indexed: 01/17/2023]
Abstract
This is the second of two studies detailing the subcortical connections of the perirhinal (PER), the postrhinal (POR) and entorhinal (EC) cortices of the rat. In the present study, we analyzed the subcortical efferents of the rat PER areas 35 and 36, POR, and the lateral and medial entorhinal areas (LEA and MEA). Anterograde tracers were injected into these five regions, and the resulting density of fiber labeling was quantified in an extensive set of subcortical structures. Density and topography of fiber labeling were quantitatively assessed in 36 subcortical areas, including olfactory structures, claustrum, amygdala nuclei, septal nuclei, basal ganglia, thalamic nuclei, and hypothalamic structures. In addition to reporting the density of labeled fibers, we incorporated a new method for quantifying the size of anterograde projections that takes into account the volume of the target subcortical structure as well as the density of fiber labeling. The PER, POR, and EC displayed unique patterns of projections to subcortical areas. Interestingly, all regions examined provided strong input to the basal ganglia, although the projections arising in the PER and LEA were stronger and more widespread. PER areas 35 and 36 exhibited similar pattern of projections with some differences. PER area 36 projects more heavily to the lateral amygdala and much more heavily to thalamic nuclei including the lateral posterior nucleus, the posterior complex, and the nucleus reuniens. Area 35 projects more heavily to olfactory structures. The LEA provides the strongest and most widespread projections to subcortical structures including all those targeted by the PER as well as the medial and posterior septal nuclei. POR shows fewer subcortical projections overall, but contributes substantial input to the lateral posterior nucleus of the thalamus. The MEA projections are even weaker. Our results suggest that the PER and LEA have greater influence over olfactory, amygdala, and septal nuclei, whereas PER area 36 and the POR have greater influence over thalamic nuclei. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kara L Agster
- Department of Neuroscience, Brown University, Providence, Rhode Island
| | - Inês Tomás Pereira
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, Rhode Island
| | - Michael P Saddoris
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, Rhode Island
| | - Rebecca D Burwell
- Department of Neuroscience, Brown University, Providence, Rhode Island.,Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, Rhode Island
| |
Collapse
|
157
|
Tomás Pereira I, Agster KL, Burwell RD. Subcortical connections of the perirhinal, postrhinal, and entorhinal cortices of the rat. I. afferents. Hippocampus 2016; 26:1189-212. [PMID: 27119220 DOI: 10.1002/hipo.22603] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 04/06/2016] [Accepted: 04/22/2016] [Indexed: 01/08/2023]
Abstract
In this study the subcortical afferents for the rat PER areas 35 and 36, POR, and the lateral and medial entorhinal areas (LEA and MEA) were characterized. We analyzed 33 retrograde tract-tracing experiments distributed across the five regions. For each experiment, we estimated the total numbers, percentages, and densities of labeled cells in 36 subcortical structures and nuclei distributed across septum, basal ganglia, claustrum, amygdala, olfactory structures, thalamus, and hypothalamus. We found that the complement of subcortical inputs differs across the five regions, especially the PER and POR. The PER receives input from the reuniens, suprageniculate, and medial geniculate thalamic nuclei as well as the amygdala. Overall, the subcortical inputs to the PER were consistent with a role in perception, multimodal processing, and the formation of associations that include the motivational significance of individual items and objects. Subcortical inputs to the POR were dominated by the dorsal thalamus, particularly the lateral posterior nucleus, a region implicated in visuospatial attention. The complement of subcortical inputs to the POR is consistent with a role in representing and monitoring the local spatial context. We also report that, in addition to the PER, the LEA and the medial band of the MEA also receive strong amygdala input. In contrast, subcortical input to the POR and the MEA lateral band includes much less amygdala input and is dominated by dorsal thalamic nuclei, particularly nuclei involved in spatial information processing. Thus, some subcortical inputs are consistent with the view that there is functional differentiation along the septotemporal axis of the hippocampus, but others provide considerable integration. Overall, we conclude that the patterns of subcortical inputs to the PER, POR, and the entorhinal LEA and MEA provide further evidence for functional differentiation in the medial temporal lobe. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Inês Tomás Pereira
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, Rhode Island, 02912
| | - Kara L Agster
- Department of Neuroscience, Brown University, Providence, Rhode Island, 02912
| | - Rebecca D Burwell
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, Rhode Island, 02912.,Department of Neuroscience, Brown University, Providence, Rhode Island, 02912
| |
Collapse
|
158
|
Machhada A, Marina N, Korsak A, Stuckey DJ, Lythgoe MF, Gourine AV. Origins of the vagal drive controlling left ventricular contractility. J Physiol 2016; 594:4017-30. [PMID: 26940639 PMCID: PMC4945717 DOI: 10.1113/jp270984] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 03/01/2016] [Indexed: 01/22/2023] Open
Abstract
Key points The strength, functional significance and origins of parasympathetic innervation of the left ventricle remain controversial. This study tested the hypothesis that parasympathetic control of left ventricular contractility is provided by vagal preganglionic neurones of the dorsal motor nucleus (DVMN). Under β‐adrenoceptor blockade combined with spinal cord (C1) transection (to remove sympathetic influences), systemic administration of atropine increased left ventricular contractility in rats anaesthetized with urethane, confirming the existence of a tonic inhibitory muscarinic influence on cardiac inotropy. Increased left ventricular contractility in anaesthetized rats was observed when DVMN neurones were silenced. Functional neuroanatomical mapping revealed that vagal preganglionic neurones that have an impact on left ventricular contractility are located in the caudal region of the left DVMN. These neurones provide functionally significant parasympathetic control of left ventricular inotropy.
Abstract The strength, functional significance and origins of direct parasympathetic innervation of the left ventricle (LV) remain controversial. In the present study we used an anaesthetized rat model to first confirm the presence of tonic inhibitory vagal influence on LV inotropy. Using genetic neuronal targeting and functional neuroanatomical mapping we tested the hypothesis that parasympathetic control of LV contractility is provided by vagal preganglionic neurones located in the dorsal motor nucleus (DVMN). It was found that under systemic β‐adrenoceptor blockade (atenolol) combined with spinal cord (C1) transection (to remove sympathetic influences), intravenous administration of atropine increases LV contractility in rats anaesthetized with urethane, but not in animals anaesthetized with pentobarbital. Increased LV contractility in rats anaesthetized with urethane was also observed when DVMN neurones targeted bilaterally to express an inhibitory Drosophila allatostatin receptor were silenced by application of an insect peptide allatostatin. Microinjections of glutamate and muscimol to activate or inhibit neuronal cell bodies in distinct locations along the rostro‐caudal extent of the left and right DVMN revealed that vagal preganglionic neurones, which have an impact on LV contractility, are located in the caudal region of the left DVMN. Changes in LV contractility were only observed when this subpopulation of DVMN neurones was activated or inhibited. These data confirm the existence of a tonic inhibitory muscarinic influence on LV contractility. Activity of a subpopulation of DVMN neurones provides functionally significant parasympathetic control of LV contractile function. The strength, functional significance and origins of parasympathetic innervation of the left ventricle remain controversial. This study tested the hypothesis that parasympathetic control of left ventricular contractility is provided by vagal preganglionic neurones of the dorsal motor nucleus (DVMN). Under β‐adrenoceptor blockade combined with spinal cord (C1) transection (to remove sympathetic influences), systemic administration of atropine increased left ventricular contractility in rats anaesthetized with urethane, confirming the existence of a tonic inhibitory muscarinic influence on cardiac inotropy. Increased left ventricular contractility in anaesthetized rats was observed when DVMN neurones were silenced. Functional neuroanatomical mapping revealed that vagal preganglionic neurones that have an impact on left ventricular contractility are located in the caudal region of the left DVMN. These neurones provide functionally significant parasympathetic control of left ventricular inotropy.
Collapse
Affiliation(s)
- Asif Machhada
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK.,UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Nephtali Marina
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Alla Korsak
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Daniel J Stuckey
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Mark F Lythgoe
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| |
Collapse
|
159
|
Early life adversities or high fat diet intake reduce cognitive function and alter BDNF signaling in adult rats: Interplay of these factors changes these effects. Int J Dev Neurosci 2016; 50:16-25. [DOI: 10.1016/j.ijdevneu.2016.03.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/01/2016] [Indexed: 01/09/2023] Open
|
160
|
Carmichael CY, Carmichael ACT, Kuwabara JT, Cunningham JT, Wainford RD. Impaired sodium-evoked paraventricular nucleus neuronal activation and blood pressure regulation in conscious Sprague-Dawley rats lacking central Gαi2 proteins. Acta Physiol (Oxf) 2016; 216:314-29. [PMID: 26412230 PMCID: PMC4764872 DOI: 10.1111/apha.12610] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 08/08/2015] [Accepted: 09/20/2015] [Indexed: 01/21/2023]
Abstract
AIM We determined the role of brain Gαi2 proteins in mediating the neural and humoral responses of conscious male Sprague-Dawley rats to acute peripheral sodium challenge. METHODS Rats pre-treated (24-h) intracerebroventricularly with a targeted oligodeoxynucleotide (ODN) (25 μg per 5 μL) to downregulate brain Gαi2 protein expression or a scrambled (SCR) control ODN were challenged with an acute sodium load (intravenous bolus 3 m NaCl; 0.14 mL per 100 g), and cardiovascular parameters were monitored for 120 min. In additional groups, hypothalamic paraventricular nucleus (PVN) Fos immunoreactivity was examined at baseline, 40, and 100 min post-sodium challenge. RESULTS In response to intravenous hypertonic saline (HS), no difference was observed in peak change in mean arterial pressure between groups. In SCR ODN pre-treated rats, arterial pressure returned to baseline by 100 min, while it remained elevated in Gαi2 ODN pre-treated rats (P < 0.05). No difference between groups was observed in sodium-evoked increases in Fos-positive magnocellular neurons or vasopressin release. V1a receptor antagonism failed to block the prolonged elevation of arterial pressure in Gαi2 ODN pre-treated rats. A significantly greater number of Fos-positive ventrolateral parvocellular, lateral parvocellular, and medial parvocellular neurons were observed in SCR vs. Gαi2 ODN pre-treated rats at 40 and 100 min post-HS challenge (P < 0.05). In SCR, but not Gαi2 ODN pre-treated rats, HS evoked suppression of plasma norepinephrine (P < 0.05). CONCLUSION This highlights Gαi2 protein signal transduction as a novel central mechanism acting to differentially influence PVN parvocellular neuronal activation, sympathetic outflow, and arterial pressure in response to acute HS, independently of actions on magnocellular neurons and vasopressin release.
Collapse
Affiliation(s)
- C. Y. Carmichael
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMAUSA
| | - A. C. T. Carmichael
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMAUSA
| | - J. T. Kuwabara
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMAUSA
| | - J. T. Cunningham
- Department of Integrative Physiology & AnatomyUniversity of North Texas Health Science CenterFort WorthTXUSA
| | - R. D. Wainford
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMAUSA
| |
Collapse
|
161
|
Gurevich EV, Gainetdinov RR, Gurevich VV. Regulation of Dopamine-Dependent Behaviors by G Protein-Coupled Receptor Kinases. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3798-1_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
162
|
Akman O, Gulcebi MI, Carcak N, Ketenci Ozatman S, Eryigit T, Moshé SL, Galanopoulou AS, Onat FY. The role of the substantia nigra pars reticulata in kindling resistance in rats with genetic absence epilepsy. Epilepsia 2015; 56:1793-802. [PMID: 26471261 DOI: 10.1111/epi.13204] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Genetic Absence Epilepsy Rats from Strasbourg (GAERS) show a resistance to secondary generalization of focal limbic seizures evoked by kindling. The substantia nigra pars reticulata (SNR) is involved in the propagation and modulation of seizures in kindling. We first examined the role of the SNRanterior and SNRposterior subregions in the resistance to the development of kindling in GAERS. Subsequently, to determine whether kindling resistance relates to differential sensitivity of γ-aminobutyric acid γ-aminobutyric acid (GABA)ergic or dopaminergic SNR neurons to kindling, we studied the effects of kindling-inducing stimulations on parvalbumin (PRV; GABAergic neuron marker) or tyrosine hydroxylase (TH; dopaminergic neuron marker) immunoreactivity (ir), respectively, in GAERS and in nonepileptic control (NEC) Wistar rats that lack kindling resistance. METHODS Adult male GAERS were implanted with a stimulation electrode in the amygdala, and bilateral injection cannulas for lidocaine or saline injection (30 min before each kindling stimulation until the animals reached three stage 5 seizures or the 22 stimulations) into the SNRanterior or SNRposterior . In another experiment, PRV-ir in SNRanterior and SNRposterior and TH-ir in SNRposterior only were densitometrically compared in GAERS-SHAM, NEC-SHAM GAERS-STIM, and NEC-STIM animals (6 kindling stimulations). RESULTS Bilateral SNRposterior infusions of lidocaine eliminated the kindling resistance and resulted in stage 5 generalized motor seizures in all kindled rats. Bilateral lidocaine infusions in the SNRanterior failed to alter the kindling resistance in GAERS. PRV-ir in the SNRposterior was unaltered in GAERS-STIM but increased in NEC-STIM group. Cellular TH-ir in the SNRposterior significantly increased by kindling stimulations in both NEC-STIM and GAERS-STIM groups. SIGNIFICANCE The kindling resistance in GAERS is mediated by the SNRposterior in a lidocaine-sensitive manner. The insensitivity to kindling stimulation of PRV-ir in SNRposterior of GAERS but not NEC rats, implicate GABAergic SNRposterior neurons in kindling resistance. In contrast, the observed stimulation-specific increase in TH-ir in the SNRposterior is unrelated to kindling resistance.
Collapse
Affiliation(s)
- Ozlem Akman
- Department of Physiology, Faculty of Medicine, Istanbul Bilim University, Istanbul, Turkey
| | - Medine I Gulcebi
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Nihan Carcak
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey
| | - Sema Ketenci Ozatman
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Marmara University, Istanbul, Turkey.,Vocational School of Health Service, Istanbul Bilim University, Istanbul, Turkey
| | - Tugba Eryigit
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Solomon L Moshé
- Laboratory of Developmental Epilepsy, Saul R. Korey Department of Neurology, Montefiore Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, U.S.A.,Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, U.S.A.,Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, U.S.A
| | - Aristea S Galanopoulou
- Laboratory of Developmental Epilepsy, Saul R. Korey Department of Neurology, Montefiore Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, U.S.A.,Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, U.S.A
| | - Filiz Yilmaz Onat
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Marmara University, Istanbul, Turkey.,Epilepsy Research Center, Marmara University, Istanbul, Turkey
| |
Collapse
|
163
|
Sabban EL, Laukova M, Alaluf LG, Olsson E, Serova LI. Locus coeruleus response to single-prolonged stress and early intervention with intranasal neuropeptide Y. J Neurochem 2015; 135:975-86. [PMID: 26333000 DOI: 10.1111/jnc.13347] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/29/2015] [Accepted: 08/15/2015] [Indexed: 01/06/2023]
Abstract
Dysregulation of the central noradrenergic system is a core feature of post-traumatic stress disorder (PTSD). Here, we examined molecular changes in locus coeruleus (LC) triggered by single-prolonged stress (SPS) PTSD model at a time when behavioral symptoms are manifested, and the effect of early intervention with intranasal neuropeptide Y (NPY). Immediately following SPS stressors, male SD rats were administered intranasal NPY (SPS/NPY) or vehicle (SPS/V). Seven days later, TH protein, but not mRNA, was elevated in LC only of the SPS/V group. Although 90% of TH positive cells expressed GR, its levels were unaltered. Compared to unstressed controls, LC of SPS/V, but not SPS/NPY, expressed less Y2 receptor mRNA with more CRHR1 mRNA in subset of animals, and elevated corticotropin-releasing hormone (CRH) in central nucleus of amygdala. Following testing for anxiety on elevated plus maze (EPM), there were significantly increased TH, DBH and NPY mRNAs in LC of SPS-treated, but not previously unstressed animals. Their levels highly correlated with each other but not with behavioral features on EPM. Thus, SPS triggers long-term noradrenergic activation and higher sensitivity to mild stressors, perhaps mediated by the up-regulation influence of amygdalar CRH input and down-regulation of Y2R presynaptic inhibition in LC. Results also demonstrate the therapeutic potential of early intervention with intranasal NPY for traumatic stress-elicited noradrenergic impairments. Single-prolonged stress (SPS)-triggered long-term changes in the locus coeruleus/norepinephrine (LC/NE) system with increased tyrosine hydroxylase (TH) protein and CRH receptor 1(CRHR1) mRNA and lower neuropeptide Y receptor 2 (Y2R) mRNA levels as well as elevated corticotropin-releasing hormone (CRH) in the central nucleus of amygdala (CeA) that were prevented by early intervention with intranasal neuropeptide Y (NPY). SPS treatment led to increased sensitivity of LC to mild stress of elevated plus maze (EPM), with elevated mRNA for NE biosynthetic enzymes in subset of animals.
Collapse
Affiliation(s)
- Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA
| | - Marcela Laukova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA.,On a long-term leave from Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lishay G Alaluf
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA
| | - Emelie Olsson
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA.,Visiting student from Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
164
|
Harada Y, Ro S, Ochiai M, Hayashi K, Hosomi E, Fujitsuka N, Hattori T, Yakabi K. Ghrelin enhancer, rikkunshito, improves postprandial gastric motor dysfunction in an experimental stress model. Neurogastroenterol Motil 2015; 27:1089-97. [PMID: 26088415 PMCID: PMC4744783 DOI: 10.1111/nmo.12588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/16/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Functional dyspepsia (FD) is one of the most common disorders of gastrointestinal (GI) diseases. However, no curable treatment is available for FD because the detailed mechanism of GI dysfunction in stressed conditions remains unclear. We aimed to clarify the association between endogenous acylated ghrelin signaling and gastric motor dysfunction and explore the possibility of a drug with ghrelin signal-enhancing action for FD treatment. METHODS Solid gastric emptying (GE) and plasma acylated ghrelin levels were evaluated in an urocortin1 (UCN1) -induced stress model. To clarify the role of acylated ghrelin on GI dysfunction in the model, exogenous acylated ghrelin, an endogenous ghrelin enhancer, rikkunshito, or an α2 -adrenergic receptor (AR) antagonist was administered. Postprandial motor function was investigated using a strain gauge force transducer in a free-moving condition. KEY RESULTS Exogenous acylated ghrelin supplementation restored UCN1-induced delayed GE. Alpha2 -AR antagonist and rikkunshito inhibited the reduction in plasma acylated ghrelin and GE in the stress model. The action of rikkunshito on delayed GE was blocked by co-administration of the ghrelin receptor antagonist. UCN1 decreased the amplitude of contraction in the antrum while increasing it in the duodenum. The motility index of the antrum but not the duodenum was significantly reduced by UCN1 treatment, which was improved by acylated ghrelin or rikkunshito. CONCLUSIONS & INFERENCES The UCN1-induced gastric motility dysfunction was mediated by abnormal acylated ghrelin dynamics. Supplementation of exogenous acylated ghrelin or enhancement of endogenous acylated ghrelin secretion by rikkunshito may be effective in treating functional GI disorders.
Collapse
Affiliation(s)
- Y. Harada
- Tsumura Research LaboratoriesTsumura & Co.IbarakiJapan
| | - S. Ro
- Department of Gastroenterology and HepatologySaitama Medical CenterSaitama Medical UniversitySaitamaJapan,Central Research LaboratoriesTeikyo University Chiba Medical CenterChibaJapan
| | - M. Ochiai
- Department of Gastroenterology and HepatologySaitama Medical CenterSaitama Medical UniversitySaitamaJapan
| | - K. Hayashi
- Department of Gastroenterology and HepatologySaitama Medical CenterSaitama Medical UniversitySaitamaJapan
| | - E. Hosomi
- Department of Gastroenterology and HepatologySaitama Medical CenterSaitama Medical UniversitySaitamaJapan
| | - N. Fujitsuka
- Tsumura Research LaboratoriesTsumura & Co.IbarakiJapan
| | - T. Hattori
- Tsumura Research LaboratoriesTsumura & Co.IbarakiJapan
| | - K. Yakabi
- Department of Gastroenterology and HepatologySaitama Medical CenterSaitama Medical UniversitySaitamaJapan
| |
Collapse
|
165
|
Bastide MF, Meissner WG, Picconi B, Fasano S, Fernagut PO, Feyder M, Francardo V, Alcacer C, Ding Y, Brambilla R, Fisone G, Jon Stoessl A, Bourdenx M, Engeln M, Navailles S, De Deurwaerdère P, Ko WKD, Simola N, Morelli M, Groc L, Rodriguez MC, Gurevich EV, Quik M, Morari M, Mellone M, Gardoni F, Tronci E, Guehl D, Tison F, Crossman AR, Kang UJ, Steece-Collier K, Fox S, Carta M, Angela Cenci M, Bézard E. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson's disease. Prog Neurobiol 2015. [PMID: 26209473 DOI: 10.1016/j.pneurobio.2015.07.002] [Citation(s) in RCA: 359] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.
Collapse
Affiliation(s)
- Matthieu F Bastide
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wassilios G Meissner
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | - Barbara Picconi
- Laboratory of Neurophysiology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Stefania Fasano
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Pierre-Olivier Fernagut
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michael Feyder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Cristina Alcacer
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yunmin Ding
- Department of Neurology, Columbia University, New York, USA
| | - Riccardo Brambilla
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre and National Parkinson Foundation Centre of Excellence, University of British Columbia, Vancouver, Canada
| | - Mathieu Bourdenx
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michel Engeln
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Sylvia Navailles
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Philippe De Deurwaerdère
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wai Kin D Ko
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Laurent Groc
- Univ. de Bordeaux, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France; CNRS, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France
| | - Maria-Cruz Rodriguez
- Department of Neurology, Hospital Universitario Donostia and Neuroscience Unit, Bio Donostia Research Institute, San Sebastian, Spain
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Maryka Quik
- Center for Health Sciences, SRI International, CA 94025, USA
| | - Michele Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Manuela Mellone
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - Dominique Guehl
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - François Tison
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | | | - Un Jung Kang
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Kathy Steece-Collier
- Michigan State University, College of Human Medicine, Department of Translational Science and Molecular Medicine & The Udall Center of Excellence in Parkinson's Disease Research, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Susan Fox
- Morton & Gloria Shulman Movement Disorders Center, Toronto Western Hospital, Toronto, Ontario M4T 2S8, Canada
| | - Manolo Carta
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Erwan Bézard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Motac Neuroscience Ltd, Manchester, UK.
| |
Collapse
|
166
|
Stoy H, Gurevich VV. How genetic errors in GPCRs affect their function: Possible therapeutic strategies. Genes Dis 2015; 2:108-132. [PMID: 26229975 PMCID: PMC4516391 DOI: 10.1016/j.gendis.2015.02.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 02/07/2015] [Indexed: 01/14/2023] Open
Abstract
Activating and inactivating mutations in numerous human G protein-coupled receptors (GPCRs) are associated with a wide range of disease phenotypes. Here we use several class A GPCRs with a particularly large set of identified disease-associated mutations, many of which were biochemically characterized, along with known GPCR structures and current models of GPCR activation, to understand the molecular mechanisms yielding pathological phenotypes. Based on this mechanistic understanding we also propose different therapeutic approaches, both conventional, using small molecule ligands, and novel, involving gene therapy.
Collapse
|
167
|
Mirshahi M, Le Marchand S. Co-purification of arrestin like proteins with alpha-enolase from bovine myocardial tissues and the possible role in heart diseases as an autoantigen. Biochem Biophys Res Commun 2015; 460:657-662. [PMID: 25824036 DOI: 10.1016/j.bbrc.2015.03.086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/16/2015] [Indexed: 11/16/2022]
Abstract
AIM Previously, we reported that visual arrestin co-purified with glycolytic enzymes. The aim of this study was to analyze the co-purification of arrestin like proteins (ALP) in bovine cardiac tissues with enolases. METHODS The soluble extract of bovine myocardial tissues from different regions such as left and right atriums and ventricles of the bovine heart (n = 3) was analyzed by ACA-34 gel filtration, immuno-affinity column, SDS-PAGE, ELISA, western blot and a sandwich immune assay for quantification of ALP and sequence analysis. RESULTS We observed that; 1) The cardiac muscle contained a 50 kDa ALP at a concentration of 751 pg/mg of soluble protein extract, 2) ALP purified, by immunoaffinity, contained alpha-enolase of 48 kDa confirmed by protein sequence analysis; 3) Cardiomyocyte cells exposed to anti arrestin and anti enolase monoclonal antibodies showed decreased proliferation in vitro, 4) High level of autoantibodies were detected by ELISA (3.57% for arrestin and 9.12% for α-enolase) in serum of patients with infarcted heart disease. CONCLUSION We suggest a possible interaction between ALP and alpha-enolases yielding a complex that may be involved in the induction of cardiac autoimmune diseases.
Collapse
Affiliation(s)
- M Mirshahi
- UMR Université Paris 7, Hôpital Lariboisière, INSERM U965, Paris, France.
| | - S Le Marchand
- UMR Université Paris 7, Hôpital Lariboisière, INSERM U965, Paris, France
| |
Collapse
|
168
|
Hansen N, Manahan-Vaughan D. Hippocampal long-term potentiation that is elicited by perforant path stimulation or that occurs in conjunction with spatial learning is tightly controlled by beta-adrenoreceptors and the locus coeruleus. Hippocampus 2015; 25:1285-98. [PMID: 25727388 PMCID: PMC6680149 DOI: 10.1002/hipo.22436] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2015] [Indexed: 11/19/2022]
Abstract
The noradrenergic system, driven by locus coeruleus (LC) activation, plays a key role in the regulating and directing of changes in hippocampal synaptic efficacy. The LC releases noradrenaline in response to novel experience and LC activation leads to an enhancement of hippocampus‐based learning, and facilitates synaptic plasticity in the form of long‐term depression (LTD) and long‐term potentiation (LTP) that occur in association with spatial learning. The predominant receptor for mediating these effects is the β‐adrenoreceptor. Interestingly, the dependency of synaptic plasticity on this receptor is different in the hippocampal subfields whereby in the CA1 in vivo, LTP, but not LTD requires β‐adrenoreceptor activation, whereas in the mossy fiber synapse LTP and LTD do not depend on this receptor. By contrast, synaptic plasticity that is facilitated by spatial learning is highly dependent on β‐adrenoreceptor activation in both hippocampal subfields. Here, we explored whether LTP induced by perforant‐path (pp) stimulation in vivo or that is facilitated by spatial learning depends on β‐adrenoreceptors. We found that under both LTP conditions, antagonising the receptors disabled the persistence of LTP. β‐adrenoreceptor‐antagonism also prevented spatial learning. Strikingly, activation of the LC before high‐frequency stimulation (HFS) of the pp prevented short‐term potentiation but not LTP, and LC stimulation after pp‐HFS‐induced depotentiation of LTP. This depotentiation was prevented by β‐adrenoreceptor‐antagonism. These data suggest that β‐adrenoreceptor‐activation, resulting from noradrenaline release from the LC during enhanced arousal and learning, comprises a mechanism whereby the duration and degree of LTP is regulated and fine tuned. This may serve to optimize the creation of a spatial memory engram by means of LTP and LTD. This process can be expected to support the special role of the dentate gyrus as a crucial subregional locus for detecting and processing novelty within the hippocampus. © 2015 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Niels Hansen
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
169
|
Gurevich VV, Gurevich EV. Arrestins: Critical Players in Trafficking of Many GPCRs. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:1-14. [PMID: 26055052 PMCID: PMC5841159 DOI: 10.1016/bs.pmbts.2015.02.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Arrestins specifically bind active phosphorylated G protein-coupled receptors (GPCRs). Receptor binding induces the release of the arrestin C-tail, which in non-visual arrestins contains high-affinity binding sites for clathrin and its adaptor AP2. Thus, serving as a physical link between the receptor and key components of the internalization machinery of the coated pit is the best-characterized function of non-visual arrestins in GPCR trafficking. However, arrestins also regulate GPCR trafficking less directly by orchestrating their ubiquitination and deubiquitination. Several reports suggest that arrestins play additional roles in receptor trafficking. Non-visual arrestins appear to be required for the recycling of internalized GPCRs, and the mechanisms of their function in this case remain to be elucidated. Moreover, visual and non-visual arrestins were shown to directly bind N-ethylmaleimide-sensitive factor, an important ATPase involved in vesicle trafficking, but neither molecular details nor the biological role of these interactions is clear. Considering how many different proteins arrestins appear to bind, we can confidently expect the elucidation of additional trafficking-related functions of these versatile signaling adaptors.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
170
|
Ahmed MR, Bychkov E, Kook S, Zurkovsky L, Dalby KN, Gurevich EV. Overexpression of GRK6 rescues L-DOPA-induced signaling abnormalities in the dopamine-depleted striatum of hemiparkinsonian rats. Exp Neurol 2015; 266:42-54. [PMID: 25687550 DOI: 10.1016/j.expneurol.2015.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 01/26/2015] [Accepted: 02/05/2015] [Indexed: 12/26/2022]
Abstract
l-DOPA therapy in Parkinson's disease often results in side effects such as l-DOPA-induced dyskinesia (LID). Our previous studies demonstrated that defective desensitization of dopamine receptors caused by decreased expression of G protein-coupled receptor kinases (GRKs) plays a role. Overexpression of GRK6, the isoform regulating dopamine receptors, in parkinsonian rats and monkeys alleviated LID and reduced LID-associated changes in gene expression. Here we show that 2-fold lentivirus-mediated overexpression of GRK6 in the dopamine-depleted striatum in rats unilaterally lesioned with 6-hydroxydopamine ameliorated supersensitive ERK response to l-DOPA challenge caused by loss of dopamine. A somewhat stronger effect of GRK6 was observed in drug-naïve than in chronically l-DOPA-treated animals. GRK6 reduced the responsiveness of p38 MAP kinase to l-DOPA challenge rendered supersensitive by dopamine depletion. The JNK MAP kinase was unaffected by loss of dopamine, chronic or acute l-DOPA, or GRK6. Overexpressed GRK6 suppressed enhanced activity of Akt in the lesioned striatum by reducing elevated phosphorylation at its major activating residue Thr(308). Finally, GRK6 reduced accumulation of ΔFosB in the lesioned striatum, the effect that paralleled a decrease in locomotor sensitization to l-DOPA in GRK6-expressing rats. The results suggest that elevated GRK6 facilitate desensitization of DA receptors, thereby normalizing of the activity of multiple signaling pathways implicated in LID. Thus, improving the regulation of dopamine receptor function via the desensitization mechanism could be an effective way of managing LID.
Collapse
Affiliation(s)
- M Rafiuddin Ahmed
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Evgeny Bychkov
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Seunghyi Kook
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lilia Zurkovsky
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kevin N Dalby
- Division of Medicinal Chemistry, University of Texas at Austin, Austin, TX 78712, USA
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
171
|
Gurevich VV, Gurevich EV. Analyzing the roles of multi-functional proteins in cells: The case of arrestins and GRKs. Crit Rev Biochem Mol Biol 2015; 50:440-452. [PMID: 26453028 PMCID: PMC4852696 DOI: 10.3109/10409238.2015.1067185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Most proteins have multiple functions. Obviously, conventional methods of manipulating the level of the protein of interest in the cell, such as over-expression, knockout or knockdown, affect all of its functions simultaneously. The key advantage of these methods is that over-expression, knockout or knockdown does not require any knowledge of the molecular mechanisms of the function(s) of the protein of interest. The disadvantage is that these approaches are inadequate to elucidate the role of an individual function of the protein in a particular cellular process. An alternative is the use of re-engineered proteins, in which a single function is eliminated or enhanced. The use of mono-functional elements of a multi-functional protein can also yield cleaner answers. This approach requires detailed knowledge of the structural basis of each function of the protein in question. Thus, a lot of preliminary structure-function work is necessary to make it possible. However, when this information is available, replacing the protein of interest with a mutant in which individual functions are modified can shed light on the biological role of those particular functions. Here, we illustrate this point using the example of protein kinases, most of which have additional non-enzymatic functions, as well as arrestins, known multi-functional signaling regulators in the cell.
Collapse
Affiliation(s)
| | - Eugenia V Gurevich
- a Department of Pharmacology , Vanderbilt University , Nashville , TN , USA
| |
Collapse
|
172
|
Hofer KT, Kandrács Á, Ulbert I, Pál I, Szabó C, Héja L, Wittner L. The hippocampal CA3 region can generate two distinct types of sharp wave-ripple complexes, in vitro. Hippocampus 2014; 25:169-86. [PMID: 25209976 DOI: 10.1002/hipo.22361] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2014] [Indexed: 11/09/2022]
Abstract
Hippocampal sharp wave-ripples (SPW-Rs) occur during slow wave sleep and behavioral immobility and are thought to play an important role in memory formation. We investigated the cellular and network properties of SPW-Rs with simultaneous laminar multielectrode and intracellular recordings in a rat hippocampal slice model, using physiological bathing medium. Spontaneous SPW-Rs were generated in the dentate gyrus (DG), CA3, and CA1 regions. These events were characterized by a local field potential gradient (LFPg) transient, increased fast oscillatory activity and increased multiple unit activity (MUA). Two types of SPW-Rs were distinguished in the CA3 region based on their different LFPg and current source density (CSD) pattern. Type 1 (T1) displayed negative LFPg transient in the pyramidal cell layer, and the associated CSD sink was confined to the proximal dendrites. Type 2 (T2) SPW-Rs were characterized by positive LFPg transient in the cell layer, and showed CSD sinks involving both the apical and basal dendrites. In both types, consistent with the somatic CSD source, only a small subset of CA3 pyramidal cells fired, most pyramidal cells were hyperpolarized, while most interneurons increased firing rate before the LFPg peak. Different neuronal populations, with different proportions of pyramidal cells and distinct subsets of interneurons were activated during T1 and T2 SPW-Rs. Activation of specific inhibitory cell subsets-with the possible leading role of perisomatic interneurons-seems to be crucial to synchronize distinct ensembles of CA3 pyramidal cells finally resulting in the expression of different SPW-R activities. This suggests that the hippocampus can generate dynamic changes in its activity stemming from the same excitatory and inhibitory circuits, and so, might provide the cellular and network basis for an input-specific and activity-dependent information transmission.
Collapse
Affiliation(s)
- Katharina T Hofer
- Department of Comparative Psychophysiology, Institute of Cognitive Neuroscience and Psychology, Research Center for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary; Department of Information Technology and Bionics, Péter Pázmány Catholic University, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
173
|
Bekinschtein P, Kent BA, Oomen CA, Clemenson GD, Gage FH, Saksida LM, Bussey TJ. Brain-derived neurotrophic factor interacts with adult-born immature cells in the dentate gyrus during consolidation of overlapping memories. Hippocampus 2014; 24:905-11. [PMID: 24825389 PMCID: PMC4312906 DOI: 10.1002/hipo.22304] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/06/2014] [Accepted: 05/09/2014] [Indexed: 12/30/2022]
Abstract
Successful memory involves not only remembering information over time but also keeping memories distinct and less confusable. The computational process for making representations of similar input patterns more distinct from each other has been referred to as "pattern separation." Although adult-born immature neurons have been implicated in this memory feature, the precise role of these neurons and associated molecules in the processing of overlapping memories is unknown. Recently, we found that brain-derived neurotrophic factor (BDNF) in the dentate gyrus is required for the encoding/consolidation of overlapping memories. In this study, we provide evidence that consolidation of these "pattern-separated" memories requires the action of BDNF on immature neurons specifically.
Collapse
Affiliation(s)
- Pedro Bekinschtein
- Department of Psychology, University of CambridgeCambridge, United Kingdom,MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, Translational and Cognitive Neuroscience Laboratory, University of CambridgeCambridge, United Kingdom,*Correspondence to: Pedro Bekinschtein, Instituto de Biología Celular y Neurociencia, Facultad de Medicina. Universidad de Buenos Aires, Buenos Aires (C1121ABG), Argentina. E-mail:
| | - Brianne A Kent
- Department of Psychology, University of CambridgeCambridge, United Kingdom,MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, Translational and Cognitive Neuroscience Laboratory, University of CambridgeCambridge, United Kingdom
| | - Charlotte A Oomen
- Department of Psychology, University of CambridgeCambridge, United Kingdom,MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, Translational and Cognitive Neuroscience Laboratory, University of CambridgeCambridge, United Kingdom
| | - Gregory D Clemenson
- Laboratory of Genetics, Salk Institute for Biological StudiesLa Jolla, California
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological StudiesLa Jolla, California
| | - Lisa M Saksida
- Department of Psychology, University of CambridgeCambridge, United Kingdom,MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, Translational and Cognitive Neuroscience Laboratory, University of CambridgeCambridge, United Kingdom
| | - Timothy J Bussey
- Department of Psychology, University of CambridgeCambridge, United Kingdom,MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, Translational and Cognitive Neuroscience Laboratory, University of CambridgeCambridge, United Kingdom
| |
Collapse
|
174
|
Zhuo Y, Vishnivetskiy SA, Zhan X, Gurevich VV, Klug CS. Identification of receptor binding-induced conformational changes in non-visual arrestins. J Biol Chem 2014; 289:20991-21002. [PMID: 24867953 PMCID: PMC4110305 DOI: 10.1074/jbc.m114.560680] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/13/2014] [Indexed: 12/25/2022] Open
Abstract
The non-visual arrestins, arrestin-2 and arrestin-3, belong to a small family of multifunctional cytosolic proteins. Non-visual arrestins interact with hundreds of G protein-coupled receptors (GPCRs) and regulate GPCR desensitization by binding active phosphorylated GPCRs and uncoupling them from heterotrimeric G proteins. Recently, non-visual arrestins have been shown to mediate G protein-independent signaling by serving as adaptors and scaffolds that assemble multiprotein complexes. By recruiting various partners, including trafficking and signaling proteins, directly to GPCRs, non-visual arrestins connect activated receptors to diverse signaling pathways. To investigate arrestin-mediated signaling, a structural understanding of arrestin activation and interaction with GPCRs is essential. Here we identified global and local conformational changes in the non-visual arrestins upon binding to the model GPCR rhodopsin. To detect conformational changes, pairs of spin labels were introduced into arrestin-2 and arrestin-3, and the interspin distances in the absence and presence of the receptor were measured by double electron electron resonance spectroscopy. Our data indicate that both non-visual arrestins undergo several conformational changes similar to arrestin-1, including the finger loop moving toward the predicted location of the receptor in the complex as well as the C-tail release upon receptor binding. The arrestin-2 results also suggest that there is no clam shell-like closure of the N- and C-domains and that the loop containing residue 136 (homolog of 139 in arrestin-1) has high flexibility in both free and receptor-bound states.
Collapse
Affiliation(s)
- Ya Zhuo
- From the Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226 and
| | - Sergey A Vishnivetskiy
- the Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Xuanzhi Zhan
- the Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Vsevolod V Gurevich
- the Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Candice S Klug
- From the Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226 and
| |
Collapse
|
175
|
You HJ, Lei J, Ye G, Fan XL, Li Q. Influence of intramuscular heat stimulation on modulation of nociception: complex role of central opioid receptors in descending facilitation and inhibition. J Physiol 2014; 592:4365-80. [PMID: 25038244 DOI: 10.1113/jphysiol.2014.275800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
It has been reported that the threshold to activate 'silent' or inactive descending facilitation of nociception is lower than that of descending inhibition. Thus, the development of pain therapy to effectively drive descending inhibition alone, without the confounding influences of facilitation is a challenge. To address this issue we investigated the effects of intramuscular stimulation with a heating-needle on spinal nociception, assessed by measuring nociceptive paw withdrawal reflex in rats. Additionally, involvement of the thalamic 'nociceptive discriminators' (thalamic mediodorsal (MD) and ventromedial (VM) nuclei), and opioid-mediated mechanisms were further explored. Descending facilitation and inhibition were elicited by 46°C noxious heating-needle stimulation, and were regulated by thalamic MD and VM nuclei, respectively. In contrast, innocuous heating-needle stimulation at a temperature of 43°C elicited descending inhibition modulated by the thalamic VM nucleus alone. Microinjection of μ/δ/κ-opioid receptor antagonists β-funaltrexamine hydrochloride/naltrindole/nor-binaltorphimine, into the VM nucleus attenuated the 46°C intramuscular heating-needle stimulation-evoked descending inhibition, whereas treatment of the MD nucleus with β-funaltrexamine hydrochloride significantly decreased the descending facilitation. By contrast, descending inhibition evoked by 43°C heating-needle stimulation was only depressed by naltrindole, as opposed to μ- and κ-opioid receptor antagonists, which failed to influence descending inhibition. The present study reveals distinct roles of μ-opioid receptors in the function of thalamic MD and VM nuclei,which exert facilitatory and inhibitory actions on nociception. Furthermore, innocuous, but not noxious, intramuscular heating-needle stimulation targeting δ-opioid receptors is suggested to be a promising avenue for the effective inhibition of pain.
Collapse
Affiliation(s)
- Hao-Jun You
- Center for Biomedical Research on Pain (CBRP), College of Medicine, Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Jing Lei
- Center for Biomedical Research on Pain (CBRP), College of Medicine, Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Gang Ye
- Department of Pain, Tongji Hospital affiliated to Shanghai Tongji University, Shanghai, 200065, P.R. China
| | - Xiao-Li Fan
- Department of Physiology, College of Medicine, Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Qiang Li
- Department of Physiology, College of Medicine, Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| |
Collapse
|
176
|
Tucci MC, Dvorkin-Gheva A, Johnson E, Cheon P, Taji L, Agarwal A, Foster J, Szechtman H. Performance of compulsive behavior in rats is not a unitary phenomenon - validation of separate functional components in compulsive checking behavior. Eur J Neurosci 2014; 40:2971-9. [PMID: 24935528 PMCID: PMC4215607 DOI: 10.1111/ejn.12652] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 05/06/2014] [Accepted: 05/08/2014] [Indexed: 11/29/2022]
Abstract
A previous analysis of the quinpirole sensitisation rat model of obsessive-compulsive disorder revealed that the behavioral phenotype of compulsive checking consists of three constitutive components – vigor of checking performance, focus on the task of checking, and satiety following a bout of checking. As confirmation of this analysis, the aim of the present study was to reconstitute, without quinpirole treatment, each of the putative components, with the expectation that these would self-assemble into compulsive checking. To reconstitute vigor and satiety, the employed treatment was a bilateral lesion of the nucleus accumbens core (NAc), as this treatment was shown previously to exaggerate these components. To reconstitute focus, the employed treatment was a low dose of the serotonin-1A receptor agonist 8-hydroxy-2-(di-n-propylamino) tetralin hydrochloride (DPAT) (0.0625 mg/kg), as high doses of this drug induce compulsive behavior and exacerbate focus. Results showed that injection of DPAT to NAc lesion rats did yield compulsive checking. Neither the drug alone nor the NAc lesion by itself produced compulsive checking. The demonstrated synthesis of compulsive checking by the combined treatment of low-dose DPAT and NAc lesion strengthened the previous fractionation of the model obsessive-compulsive disorder phenotype into three constitutive components, and suggested a role for serotonin-1A receptors outside the NAc in enhanced focus on the task of checking.
Collapse
Affiliation(s)
- Mark C Tucci
- Department of Psychiatry and Behavioural Neurosciences, Health Science Centre, McMaster University, Room 4N82, 1280 Main Street West, Hamilton, ON, Canada, L8S 4K1
| | | | | | | | | | | | | | | |
Collapse
|
177
|
Marco A, Kisliouk T, Tabachnik T, Meiri N, Weller A. Overweight and CpG methylation of the Pomc promoter in offspring of high-fat-diet-fed dams are not "reprogrammed" by regular chow diet in rats. FASEB J 2014; 28:4148-57. [PMID: 24928196 DOI: 10.1096/fj.14-255620] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 06/02/2014] [Indexed: 12/16/2022]
Abstract
This study aimed to determine whether epigenetic malprogramming induced by high-fat diet (HFD) has an obesogenic effect on nonmated and mated female rats and their offspring. Further, it aimed to reprogram offspring's epigenetic malprogramming and phenotype by providing normal diet after weaning. Body weight (BW) was measured, and plasma and hypothalamic arcuate nuclei were collected for analysis of hormones, mRNA, and DNA CpG methylation of the promoter of Pomc, a key factor in control of food intake. In nonmated females, HFD decreased Pomc/leptin ratio by ∼38%. This finding was associated with Pomc promoter hypermethylation. While heavier during pregnancy, during lactation HFD dams showed sharper BW decrease (2.5-fold) and loss of Pomc promoter hypermethylation. Moreover, their weight loss was correlated with demethylation (r=-0.707) and with gadd45b mRNA expression levels (r=0.905). Even though offspring of HFD dams ate standard chow from weaning, they displayed increased BW, Pomc promoter hypermethylation, and vulnerability to HFD challenge (3-fold kilocalorie intake increase). These findings demonstrate a long-term effect of maternal HFD on CpG methylation of the Pomc promoter in the offspring, which was not reprogrammed by standard chow from weaning. Further, the results suggest a possible mechanism of demethylation of the Pomc promoter following pregnancy and lactation.
Collapse
Affiliation(s)
- Asaf Marco
- Faculty of Life Sciences, Gonda Brain Research Center, and
| | - Tatiana Kisliouk
- Institute of Animal Science, Agricultural Research Organization, Volcani Center, Bet Dagan, Israel
| | | | - Noam Meiri
- Institute of Animal Science, Agricultural Research Organization, Volcani Center, Bet Dagan, Israel
| | - Aron Weller
- Gonda Brain Research Center, and Department of Psychology, Bar Ilan University, Ramat-Gan, Israel; and
| |
Collapse
|
178
|
Zhan X, Perez A, Gimenez LE, Vishnivetskiy SA, Gurevich VV. Arrestin-3 binds the MAP kinase JNK3α2 via multiple sites on both domains. Cell Signal 2014; 26:766-776. [PMID: 24412749 PMCID: PMC3936466 DOI: 10.1016/j.cellsig.2014.01.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/18/2013] [Accepted: 01/02/2014] [Indexed: 01/14/2023]
Abstract
Although arrestins bind dozens of non-receptor partners, the interaction sites for most signaling proteins remain unknown. Here we report the identification of arrestin-3 elements involved in binding MAP kinase JNK3α2. Using purified JNK3α2 and MBP fusions containing separated arrestin-3 domains and peptides exposed on the non-receptor-binding surface of arrestin-3 we showed that both domains bind JNK3α2 and identified one element on the N-domain and two on the C-domain that directly interact with JNK3α2. Using in vitro competition we confirmed that JNK3α2 engages identified N-domain element and one of the C-domain peptides in the full-length arrestin-3. The 25-amino acid N-domain element has the highest affinity for JNK3α2, suggesting that it is the key site for JNK3α2 docking. The identification of elements involved in protein-protein interactions paves the way to targeted redesign of signaling proteins to modulate cell signaling in desired ways. The tools and methods developed here to elucidate the molecular mechanism of arrestin-3 interactions with JNK3α2 are suitable for mapping of arrestin-3 sites involved in interactions with other partners.
Collapse
Affiliation(s)
- Xuanzhi Zhan
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Alejandro Perez
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Luis E Gimenez
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
179
|
Gurevich VV, Gurevich EV. Extensive shape shifting underlies functional versatility of arrestins. Curr Opin Cell Biol 2014; 27:1-9. [PMID: 24680424 PMCID: PMC3971385 DOI: 10.1016/j.ceb.2013.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/20/2013] [Accepted: 10/23/2013] [Indexed: 12/24/2022]
Abstract
Among four vertebrate arrestins, only two are ubiquitously expressed. Arrestins specifically bind active phosphorylated G protein-coupled receptors (GPCRs), thereby precluding further G protein activation. Recent discoveries suggest that the formation of the arrestin-receptor complex initiates the second round of signaling with comparable biological importance. Despite having virtually no recognizable sequence motifs known to mediate protein-protein interactions, arrestins bind a surprising variety of signaling proteins with mind-boggling range of functional consequences. High conformational flexibility allows arrestins to show many distinct 'faces' to the world, which allows these relatively small ∼45kDa proteins to bind various partners under different physiological conditions, organizing multi-protein signaling complexes and localizing them to distinct subcellular compartments.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
180
|
Zaretsky DV, Zaretskaia MV, Durant PJ, Rusyniak DE. Inhibition of the dorsomedial hypothalamus, but not the medullary raphe pallidus, decreases hyperthermia and mortality from MDMA given in a warm environment. Pharmacol Res Perspect 2014; 2:e00031. [PMID: 24765530 PMCID: PMC3994179 DOI: 10.1002/prp2.31] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The central mechanisms through which MDMA mediates life-threatening hyperthermia when taken in a warm environment are not well described. It is assumed that MDMA alters normal thermoregulatory circuits resulting in increased heat production through interscapular brown adipose tissue (iBAT) and decreased heat dissipation through cutaneous vasoconstriction. We studied the role of the dorsomedial hypothalamus (DMH) and medullary raphe pallidus (mRPa) in mediating iBAT, tail blood flow, and locomotor effects produced by MDMA. Rats were instrumented with guide cannulas targeting either the DMH or the mRPa-brain regions involved in regulating iBAT and cutaneous vascular beds. In all animals, core temperature and locomotion were recorded with surgically implanted telemetric transmitters; and additionally either iBAT temperature (via telemetric transmitter) or tail artery blood flow (via tail artery Doppler cuff) were also recorded. Animals were placed in an environmental chamber at 32°C and microinjected with either control or the GABA agonist muscimol (80pmol) followed by an intravenous injection of saline or MDMA (7.5 mg kg-1). To prevent undue suffering, a core temperature of 41°C was chosen as the surrogate marker of mortality. Inhibition of the DMH, but not the mRPa, prevented mortality and attenuated hyperthermia and locomotion. Inhibition of either the DMH or the mRPa did not affect iBAT temperature increases or tail blood flow decreases. While MDMA increases iBAT thermogenesis and decreases heat dissipation through cutaneous vasoconstriction, thermoregulatory brain regions known to mediate these effects are not involved. Rather, the finding that inhibiting the DMH decreases both locomotion and body temperature suggests that locomotion may be a key central contributor to MDMA-evoked hyperthermia.
Collapse
Affiliation(s)
- Dmitry V Zaretsky
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Maria V Zaretskaia
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Pamela J Durant
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Daniel E Rusyniak
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA ; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
181
|
Çarçak N, Zheng T, Ali I, Abdullah A, French C, Powell KL, Jones NC, van Raay L, Rind G, Onat F, O'Brien TJ. The effect of amygdala kindling on neuronal firing patterns in the lateral thalamus in the GAERS model of absence epilepsy. Epilepsia 2014; 55:654-665. [DOI: 10.1111/epi.12592] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Nihan Çarçak
- Department of Pharmacology; Faculty of Pharmacy; Istanbul University; Istanbul Turkey
| | - Thomas Zheng
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
| | - Idrish Ali
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
| | - Ahmad Abdullah
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
| | - Chris French
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
- Department of Neurology; Royal Melbourne Hospital; Melbourne Vic. Australia
| | - Kim L. Powell
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
| | - Nigel C. Jones
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
| | - Leena van Raay
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
| | - Gil Rind
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
| | - Filiz Onat
- Department of Pharmacology and Clinical Pharmacology; Marmara University School of Medicine; Istanbul Turkey
| | - Terence J. O'Brien
- Department of Medicine; Royal Melbourne Hospital; University of Melbourne; Melbourne Vic. Australia
- Department of Neurology; Royal Melbourne Hospital; Melbourne Vic. Australia
| |
Collapse
|
182
|
Roberts MD, Toedebusch RG, Wells KD, Company JM, Brown JD, Cruthirds CL, Heese AJ, Zhu C, Rottinghaus GE, Childs TE, Booth FW. Nucleus accumbens neuronal maturation differences in young rats bred for low versus high voluntary running behaviour. J Physiol 2014; 592:2119-35. [PMID: 24665095 DOI: 10.1113/jphysiol.2013.268805] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We compared the nucleus accumbens (NAc) transcriptomes of generation 8 (G8), 34-day-old rats selectively bred for low (LVR) versus high voluntary running (HVR) behaviours in rats that never ran (LVR(non-run) and HVR(non-run)), as well as in rats after 6 days of voluntary wheel running (LVR(run) and HVR(run)). In addition, the NAc transcriptome of wild-type Wistar rats was compared. The purpose of this transcriptomics approach was to generate testable hypotheses as to possible NAc features that may be contributing to running motivation differences between lines. Ingenuity Pathway Analysis and Gene Ontology analyses suggested that 'cell cycle'-related transcripts and the running-induced plasticity of dopamine-related transcripts were lower in LVR versus HVR rats. From these data, a hypothesis was generated that LVR rats might have less NAc neuron maturation than HVR rats. Follow-up immunohistochemistry in G9-10 LVR(non-run) rats suggested that the LVR line inherently possessed fewer mature medium spiny (Darpp-32-positive) neurons (P < 0.001) and fewer immature (Dcx-positive) neurons (P < 0.001) than their G9-10 HVR counterparts. However, voluntary running wheel access in our G9-10 LVRs uniquely increased their Darpp-32-positive and Dcx-positive neuron densities. In summary, NAc cellularity differences and/or the lack of running-induced plasticity in dopamine signalling-related transcripts may contribute to low voluntary running motivation in LVR rats.
Collapse
Affiliation(s)
- Michael D Roberts
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Ryan G Toedebusch
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Kevin D Wells
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Joseph M Company
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Jacob D Brown
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Clayton L Cruthirds
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Alexander J Heese
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Conan Zhu
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - George E Rottinghaus
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Thomas E Childs
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Frank W Booth
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
183
|
Jeggo R, Zhao FY, Spanswick D. Electrophysiological Techniques for Studying Synaptic Activity In Vivo. CURRENT PROTOCOLS IN PHARMACOLOGY 2014; 64:11.11.1-17. [PMID: 26344209 DOI: 10.1002/0471141755.ph1111s64] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Understanding the physiology, pharmacology, and plasticity associated with synaptic function is a key goal of neuroscience research and is fundamental to identifying the processes involved in the development and manifestation of neurological disease. A diverse range of electrophysiological methodologies are used to study synaptic function. Described in this unit is a technique for recording electrical activity from a single component of the central nervous system that is used to investigate pre- and post-synaptic elements of synaptic function. A strength of this technique is that it can be used on live animals, although the effect of anesthesia must be taken into consideration when interpreting the results. This methodology can be employed not only in naïve animals for studying normal physiological synaptic function, but also in a variety of disease models, including transgenic animals, to examine dysfunctional synaptic plasticity associated with neurological pathologies.
Collapse
Affiliation(s)
- Ross Jeggo
- Neurosolutions Limited, University of Warwick Medical School, Coventry, United Kingdom
| | - Fei-Yue Zhao
- Neurosolutions Limited, University of Warwick Medical School, Coventry, United Kingdom
| | - David Spanswick
- The Institute For Genomic Research, Rockville, Maryland.,Department of Physiology, Monash University, Clayton, Victoria, Australia.,University of Warwick Medical School, Coventry, United Kingdom
| |
Collapse
|
184
|
Belchior H, Lopes-Dos-Santos V, Tort ABL, Ribeiro S. Increase in hippocampal theta oscillations during spatial decision making. Hippocampus 2014; 24:693-702. [PMID: 24520011 PMCID: PMC4229028 DOI: 10.1002/hipo.22260] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 01/31/2014] [Accepted: 02/04/2014] [Indexed: 01/01/2023]
Abstract
The processing of spatial and mnemonic information is believed to depend on hippocampal theta oscillations (5-12 Hz). However, in rats both the power and the frequency of the theta rhythm are modulated by locomotor activity, which is a major confounding factor when estimating its cognitive correlates. Previous studies have suggested that hippocampal theta oscillations support decision-making processes. In this study, we investigated to what extent spatial decision making modulates hippocampal theta oscillations when controlling for variations in locomotion speed. We recorded local field potentials from the CA1 region of rats while animals had to choose one arm to enter for reward (goal) in a four-arm radial maze. We observed prominent theta oscillations during the decision-making period of the task, which occurred in the center of the maze before animals deliberately ran through an arm toward goal location. In speed-controlled analyses, theta power and frequency were higher during the decision period when compared to either an intertrial delay period (also at the maze center), or to the period of running toward goal location. In addition, theta activity was higher during decision periods preceding correct choices than during decision periods preceding incorrect choices. Altogether, our data support a cognitive function for the hippocampal theta rhythm in spatial decision making.
Collapse
Affiliation(s)
- Hindiael Belchior
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil; Psychobiology Graduate Program, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | | | | |
Collapse
|
185
|
Gurevich VV, Song X, Vishnivetskiy SA, Gurevich EV. Enhanced phosphorylation-independent arrestins and gene therapy. Handb Exp Pharmacol 2014; 219:133-152. [PMID: 24292828 PMCID: PMC4516159 DOI: 10.1007/978-3-642-41199-1_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
A variety of heritable and acquired disorders is associated with excessive signaling by mutant or overstimulated GPCRs. Since any conceivable treatment of diseases caused by gain-of-function mutations requires gene transfer, one possible approach is functional compensation. Several structurally distinct forms of enhanced arrestins that bind phosphorylated and even non-phosphorylated active GPCRs with much higher affinity than parental wild-type proteins have the ability to dampen the signaling by hyperactive GPCR, pushing the balance closer to normal. In vivo this approach was so far tested only in rod photoreceptors deficient in rhodopsin phosphorylation, where enhanced arrestin improved the morphology and light sensitivity of rods, prolonged their survival, and accelerated photoresponse recovery. Considering that rods harbor the fastest, as well as the most demanding and sensitive GPCR-driven signaling cascade, even partial success of functional compensation of defect in rhodopsin phosphorylation by enhanced arrestin demonstrates the feasibility of this strategy and its therapeutic potential.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, Nashville, TN, 37232, USA,
| | | | | | | |
Collapse
|
186
|
Gimenez LE, Vishnivetskiy SA, Gurevich VV. Targeting individual GPCRs with redesigned nonvisual arrestins. Handb Exp Pharmacol 2014; 219:153-70. [PMID: 24292829 DOI: 10.1007/978-3-642-41199-1_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Numerous human diseases are caused by excessive signaling of mutant G protein-coupled receptors (GPCRs) or receptors that are overstimulated due to upstream signaling imbalances. The feasibility of functional compensation by arrestins with enhanced ability to quench receptor signaling was recently tested in the visual system. The results showed that even in this extremely demanding situation of rods that have no ability to phosphorylate rhodopsin, enhanced arrestin improved rod morphology, light sensitivity, survival, and accelerated photoresponse recovery. Structurally distinct enhanced mutants of arrestins that bind phosphorylated and non-phosphorylated active GPCRs with much higher affinity than parental wild-type (WT) proteins have been constructed. These "super-arrestins" are likely to have the power to dampen the signaling by hyperactive GPCRs. However, most cells express 5-20 GPCR subtypes, only one of which would be overactive, while nonvisual arrestins are remarkably promiscuous, binding hundreds of different GPCRs. Thus, to be therapeutically useful, enhanced versions of nonvisual arrestins must be made fairly specific for particular receptors. Recent identification of very few arrestin residues as key receptor discriminators paves the way to the construction of receptor subtype-specific nonvisual arrestins.
Collapse
Affiliation(s)
- Luis E Gimenez
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, Nashville, TN, 37232, USA,
| | | | | |
Collapse
|
187
|
Kook S, Zhan X, Kaoud TS, Dalby KN, Gurevich VV, Gurevich EV. Arrestin-3 binds c-Jun N-terminal kinase 1 (JNK1) and JNK2 and facilitates the activation of these ubiquitous JNK isoforms in cells via scaffolding. J Biol Chem 2013; 288:37332-37342. [PMID: 24257757 PMCID: PMC3873585 DOI: 10.1074/jbc.m113.510412] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 11/07/2013] [Indexed: 12/29/2022] Open
Abstract
Non-visual arrestins scaffold mitogen-activated protein kinase (MAPK) cascades. The c-Jun N-terminal kinases (JNKs) are members of MAPK family. Arrestin-3 has been shown to enhance the activation of JNK3, which is expressed mainly in neurons, heart, and testes, in contrast to ubiquitous JNK1 and JNK2. Although all JNKs are activated by MKK4 and MKK7, both of which bind arrestin-3, the ability of arrestin-3 to facilitate the activation of JNK1 and JNK2 has never been reported. Using purified proteins we found that arrestin-3 directly binds JNK1α1 and JNK2α2, interacting with the latter comparably to JNK3α2. Phosphorylation of purified JNK1α1 and JNK2α2 by MKK4 or MKK7 is increased by arrestin-3. Endogenous arrestin-3 interacted with endogenous JNK1/2 in different cell types. Arrestin-3 also enhanced phosphorylation of endogenous JNK1/2 in intact cells upon expression of upstream kinases ASK1, MKK4, or MKK7. We observed a biphasic effect of arrestin-3 concentrations on phosphorylation of JNK1α1 and JNK2α2 both in vitro and in vivo. Thus, arrestin-3 acts as a scaffold, facilitating JNK1α1 and JNK2α2 phosphorylation by MKK4 and MKK7 via bringing JNKs and their activators together. The data suggest that arrestin-3 modulates the activity of ubiquitous JNK1 and JNK2 in non-neuronal cells, impacting the signaling pathway that regulates their proliferation and survival.
Collapse
Affiliation(s)
- Seunghyi Kook
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - Xuanzhi Zhan
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - Tamer S. Kaoud
- Faculty of Pharmacy, Minia University, Minia 61519, Egypt, and
- Division of Medicinal Chemistry, The University of Texas, Austin, Texas 78712
| | - Kevin N. Dalby
- Division of Medicinal Chemistry, The University of Texas, Austin, Texas 78712
| | - Vsevolod V. Gurevich
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - Eugenia V. Gurevich
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| |
Collapse
|
188
|
Effects of undernourishment, recurrent seizures and enriched environment during early life in hippocampal morphology. Int J Dev Neurosci 2013; 33:81-7. [DOI: 10.1016/j.ijdevneu.2013.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/13/2013] [Accepted: 12/13/2013] [Indexed: 11/21/2022] Open
|
189
|
Hopkins SC, Campbell UC, Heffernan MLR, Spear KL, Jeggo RD, Spanswick DC, Varney MA, Large TH. Effects of D-amino acid oxidase inhibition on memory performance and long-term potentiation in vivo. Pharmacol Res Perspect 2013; 1:e00007. [PMID: 25505561 PMCID: PMC4184572 DOI: 10.1002/prp2.7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/16/2013] [Indexed: 12/11/2022] Open
Abstract
N-methyl-d-aspartate receptor (NMDAR) activation can initiate changes in synaptic strength, evident as long-term potentiation (LTP), and is a key molecular correlate of memory formation. Inhibition of d-amino acid oxidase (DAAO) may increase NMDAR activity by regulating d-serine concentrations, but which neuronal and behavioral effects are influenced by DAAO inhibition remain elusive. In anesthetized rats, extracellular field excitatory postsynaptic potentials (fEPSPs) were recorded before and after a theta frequency burst stimulation (TBS) of the Schaffer collateral pathway of the CA1 region in the hippocampus. Memory performance was assessed after training with tests of contextual fear conditioning (FC, mice) and novel object recognition (NOR, rats). Oral administration of 3, 10, and 30 mg/kg 4H-furo[3,2-b]pyrrole-5-carboxylic acid (SUN) produced dose-related and steady increases of cerebellum d-serine in rats and mice, indicative of lasting inhibition of central DAAO. SUN administered 2 h prior to training improved contextual fear conditioning in mice and novel object recognition memory in rats when tested 24 h after training. In anesthetized rats, LTP was established proportional to the number of TBS trains. d-cycloserine (DCS) was used to identify a submaximal level of LTP (5× TBS) that responded to NMDA receptor activation; SUN administered at 10 mg/kg 3-4 h prior to testing similarly increased in vivo LTP levels compared to vehicle control animals. Interestingly, in vivo administration of DCS also increased brain d-serine concentrations. These results indicate that DAAO inhibition increased NMDAR-related synaptic plasticity during phases of post training memory consolidation to improve memory performance in hippocampal-dependent behavioral tests.
Collapse
Affiliation(s)
| | | | | | - Kerry L Spear
- Sunovion Pharmaceuticals IncMarlborough, Massachusetts
| | | | - David C Spanswick
- Neurosolutions Ltd.Coventry, U.K
- Department of Physiology, Monash UniversityClayton, Victoria, Australia
- Warwick Medical School, University of WarwickCoventry, U.K
| | - Mark A Varney
- Sunovion Pharmaceuticals IncMarlborough, Massachusetts
| | | |
Collapse
|
190
|
Large scale expression and purification of mouse melanopsin-L in the baculovirus expression system. Protein Expr Purif 2013; 91:134-46. [PMID: 23921072 DOI: 10.1016/j.pep.2013.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/14/2013] [Accepted: 07/17/2013] [Indexed: 11/22/2022]
Abstract
Melanopsin is the mammalian photopigment that primarily mediates non-visual photoregulated physiology. So far, this photopigment is poorly characterized with respect to structure and function. Here, we report large-scale production and purification of the intact long isoform of mouse melanopsin (melanopsin-L) using the baculovirus/insect cell expression system. Exploiting the baculoviral GP67 signal peptide, we obtained expression levels that varied between 10-30pmol/10(6)cells, equivalent to 2-5mg/L. This could be further enhanced using DMSO as a chemical chaperone. LC-MS analysis confirmed that full-length melanopsin-L was expressed and demonstrated that the majority of the expressed protein was N-glycosylated at Asn(30) and Asn(34). Other posttranslational modifications were not yet detected. Purification was achieved exploiting a C-terminal deca-histag, realizing a purification factor of several hundred-fold. The final recovery of purified melanopsin-L averaged 2.5% of the starting material. This was mainly due to low extraction yields, probably since most of the protein was present as the apoprotein. The spectral data we obtained agree with an absorbance maximum in the 460-500nm wavelength region and a significant red-shift upon illumination. This is the first report on expression and purification of full length melanopsin-L at a scale that can easily be further amplified.
Collapse
|
191
|
Pelloux Y, Murray JE, Everitt BJ. Differential roles of the prefrontal cortical subregions and basolateral amygdala in compulsive cocaine seeking and relapse after voluntary abstinence in rats. Eur J Neurosci 2013; 38:3018-26. [PMID: 23815783 PMCID: PMC3910160 DOI: 10.1111/ejn.12289] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 05/31/2013] [Indexed: 01/17/2023]
Abstract
Compulsive drug use and a persistent vulnerability to relapse are key features of addiction. Imaging studies have suggested that these features may result from deficits in prefrontal cortical structure and function, and thereby impaired top-down inhibitory control over limbic-striatal mechanisms of drug-seeking behaviour. We tested the hypothesis that selective damage to distinct subregions of the prefrontal cortex, or to the amygdala, after a short history of cocaine taking would: (i) result in compulsive cocaine seeking at a time when it would not usually be displayed; or (ii) facilitate relapse to drug seeking after abstinence. Rats with selective, bilateral excitotoxic lesions of the basolateral amygdala or anterior cingulate, prelimbic, infralimbic, orbitofrontal or anterior insular cortices were trained to self-administer cocaine under a seeking-taking chained schedule. Intermittent mild footshock punishment of the cocaine-seeking response was then introduced. No prefrontal cortical lesion affected the ability of rats to withhold their seeking responses. However, rats with lesions to the basolateral amygdala increased their cocaine-seeking responses under punishment and were impaired in their acquisition of conditioned fear. Following a 7-day abstinence period, rats were re-exposed to the drug-seeking environment for assessment of relapse in the absence of punishment or cocaine. Rats with prelimbic cortex lesions showed decreased seeking responses during relapse, whereas those with anterior insular cortex lesions showed an increase. Combined, these results show that acute impairment of prefrontal cortical function does not result in compulsive cocaine seeking after a short history of self-administering cocaine, but further implicates subregions of the prefrontal cortex in relapse.
Collapse
Affiliation(s)
- Yann Pelloux
- Institut de Neuroscience de la Timone, UMR 7289 CNRS, Université d'Aix-Marseille, 27 Bld Jean Moulin, 13385 Marseille, France.
| | - Jennifer E Murray
- Department of Psychology, University of Cambridge, Downing St, CB2 3EB, Cambridge, UK.,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing St, CB2 3EB, Cambridge, UK
| | - Barry J Everitt
- Department of Psychology, University of Cambridge, Downing St, CB2 3EB, Cambridge, UK.,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing St, CB2 3EB, Cambridge, UK
| |
Collapse
|
192
|
Vishnivetskiy SA, Baameur F, Findley KR, Gurevich VV. Critical role of the central 139-loop in stability and binding selectivity of arrestin-1. J Biol Chem 2013; 288:11741-11750. [PMID: 23476014 PMCID: PMC3636863 DOI: 10.1074/jbc.m113.450031] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 02/28/2013] [Indexed: 11/06/2022] Open
Abstract
Arrestin-1 selectively binds active phosphorylated rhodopsin (P-Rh*), demonstrating much lower affinity for inactive phosphorylated (P-Rh) and unphosphorylated active (Rh*) forms. Receptor interaction induces significant conformational changes in arrestin-1, which include large movement of the previously neglected 139-loop in the center of the receptor binding surface, away from the incoming receptor. To elucidate the functional role of this loop, in mouse arrestin-1 we introduced deletions of variable lengths and made several substitutions of Lys-142 in it and Asp-72 in the adjacent loop. Several mutants with perturbations in the 139-loop demonstrate increased binding to P-Rh*, dark P-Rh, Rh*, and phospho-opsin. Enhanced binding of arrestin-1 mutants to non-preferred forms of rhodopsin correlates with decreased thermal stability. The 139-loop perturbations increase P-Rh* binding of arrestin-1 at low temperatures and further change its binding profile on the background of 3A mutant, where the C-tail is detached from the body of the molecule by triple alanine substitution. Thus, the 139-loop stabilizes basal conformation of arrestin-1 and acts as a brake, preventing its binding to non-preferred forms of rhodopsin. Conservation of this loop in other subtypes suggests that it has the same function in all members of the arrestin family.
Collapse
Affiliation(s)
| | - Faiza Baameur
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - Kristen R. Findley
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - Vsevolod V. Gurevich
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| |
Collapse
|
193
|
Bajic D, Commons KG, Soriano SG. Morphine-enhanced apoptosis in selective brain regions of neonatal rats. Int J Dev Neurosci 2013; 31:258-66. [PMID: 23499314 DOI: 10.1016/j.ijdevneu.2013.02.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 02/09/2013] [Accepted: 02/28/2013] [Indexed: 12/12/2022] Open
Abstract
Prolonged neonatal opioid exposure has been associated with: antinociceptive tolerance, long-term neurodevelopmental delay, cognitive, and motor impairment. Morphine has also been shown to induce apoptotic cell death in vitro studies, but its in vivo effect in developing rat brain is unknown. Thus, we hypothesized that prolongued morphine administration in neonatal rats in a model of antinociceptive tolerance and dependence is associated with increased neuroapoptosis. We analyzed neonatal rats from the following groups (1) naïve group (n=6); (2) control group (normal saline (NS), n=5), and (3) morphine group (n=8). Morphine sulfate or equal volume of NS was injected subcutaneously twice daily for 6½ days starting on postnatal day (PD) 1. Development of antinociceptive tolerance was previously confirmed by Hot Plate test on the 7th day. Evidence of neuronal and glial apoptosis was determined by cleaved caspase-3 immunofluorescence combined with specific markers. At PD7, morphine administration after 6½ days significantly increased the density of apoptotic cells in the cortex and amygdala, but not in the hippocampus, hypothalamus, or periaqueductal gray. Apoptotic cells exhibited morphology analogous to neurons. Irrespective of the treatment, only a very few individual microglia but not astrocytes were caspase-3 positive. In summary, repeated morphine administration in neonatal rats (PD1-7) is associated with increased supraspinal apoptosis in distinct anatomical regions known to be important for sensory (cortex) and emotional memory processing (amygdala). Brain regions important for learning (hippocampus), and autonomic and nociceptive processing (hypothalamus and periaqueductal gray) were not affected. Lack of widespread glial apoptosis or robust glial activation following repeated morphine administration suggests that glia might not be affected by chronic morphine at this early age. Future studies should investigate long-term behavioral sequelae of demonstrated enhanced apoptosis associated with prolonged morphine administration in a neonatal rat model.
Collapse
Affiliation(s)
- Dusica Bajic
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, 300 Longwood Avenue, Bader 3, Boston, MA 02115, USA.
| | | | | |
Collapse
|
194
|
Bustamante C, Henríquez R, Medina F, Reinoso C, Vargas R, Pascual R. Maternal exercise during pregnancy ameliorates the postnatal neuronal impairments induced by prenatal restraint stress in mice. Int J Dev Neurosci 2013; 31:267-73. [PMID: 23466414 DOI: 10.1016/j.ijdevneu.2013.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 02/18/2013] [Accepted: 02/18/2013] [Indexed: 12/16/2022] Open
Abstract
Clinical and preclinical studies have demonstrated that prenatal stress (PS) induces neuronal and behavioral disturbances in the offspring. In the present study, we determined whether maternal voluntary wheel running (VWR) during pregnancy could reverse the putative deleterious effects of PS on the neurodevelopment and behavior of the offspring. Pregnant CF-1 mice were randomly assigned to control, restraint stressed or restraint stressed+VWR groups. Dams of the stressed group were subjected to restraint stress between gestational days 14 and delivery, while control pregnant dams remained undisturbed in their home cages. Dams of the restraint stressed+VWR group were subjected to exercise between gestational days 1 and 17. On postnatal day 23 (P23), male pups were assigned to one of the following experimental groups: mice born from control dams, stressed dams or stressed+VWR dams. Locomotor behavior and pyramidal neuronal morphology were evaluated at P23. Animals were then sacrificed, and Golgi-impregnated pyramidal neurons of the parietal cortex were morphometrically analyzed. Here, we present two major findings: first, PS produced significantly diminished dendritic growth of parietal neurons without altered locomotor behavior of the offspring; and second, maternal VWR significantly offset morphological impairments.
Collapse
Affiliation(s)
- Carlos Bustamante
- Laboratorio de Neurociencias, Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Chile.
| | | | | | | | | | | |
Collapse
|
195
|
Wilson DIG, Langston RF, Schlesiger MI, Wagner M, Watanabe S, Ainge JA. Lateral entorhinal cortex is critical for novel object-context recognition. Hippocampus 2013; 23:352-66. [PMID: 23389958 PMCID: PMC3648979 DOI: 10.1002/hipo.22095] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2013] [Indexed: 11/10/2022]
Abstract
Episodic memory incorporates information about specific events or occasions including spatial locations and the contextual features of the environment in which the event took place. It has been modeled in rats using spontaneous exploration of novel configurations of objects, their locations, and the contexts in which they are presented. While we have a detailed understanding of how spatial location is processed in the brain relatively little is known about where the nonspatial contextual components of episodic memory are processed. Initial experiments measured c-fos expression during an object-context recognition (OCR) task to examine which networks within the brain process contextual features of an event. Increased c-fos expression was found in the lateral entorhinal cortex (LEC; a major hippocampal afferent) during OCR relative to control conditions. In a subsequent experiment it was demonstrated that rats with lesions of LEC were unable to recognize object-context associations yet showed normal object recognition and normal context recognition. These data suggest that contextual features of the environment are integrated with object identity in LEC and demonstrate that recognition of such object-context associations requires the LEC. This is consistent with the suggestion that contextual features of an event are processed in LEC and that this information is combined with spatial information from medial entorhinal cortex to form episodic memory in the hippocampus. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David I G Wilson
- School of Psychology, University of St Andrews, St Mary's Quad, St Andrews, Fife, United Kingdom
| | | | | | | | | | | |
Collapse
|
196
|
Vishnivetskiy SA, Chen Q, Palazzo MC, Brooks EK, Altenbach C, Iverson TM, Hubbell WL, Gurevich VV. Engineering visual arrestin-1 with special functional characteristics. J Biol Chem 2013; 288:3394-3405. [PMID: 23250748 PMCID: PMC3561558 DOI: 10.1074/jbc.m112.445437] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Indexed: 01/31/2023] Open
Abstract
Arrestin-1 preferentially binds active phosphorylated rhodopsin. Previously, a mutant with enhanced binding to unphosphorylated active rhodopsin (Rh*) was shown to partially compensate for lack of rhodopsin phosphorylation in vivo. Here we showed that reengineering of the receptor binding surface of arrestin-1 further improves the binding to Rh* while preserving protein stability. In mammals, arrestin-1 readily self-associates at physiological concentrations. The biological role of this phenomenon can only be elucidated by replacing wild type arrestin-1 in living animals with a non-oligomerizing mutant retaining all other functions. We demonstrate that constitutively monomeric forms of arrestin-1 are sufficiently stable for in vivo expression. We also tested the idea that individual functions of arrestin-1 can be independently manipulated to generate mutants with the desired combinations of functional characteristics. Here we showed that this approach is feasible; stable forms of arrestin-1 with high Rh* binding can be generated with or without the ability to self-associate. These novel molecular tools open the possibility of testing of the biological role of arrestin-1 self-association and pave the way to elucidation of full potential of compensational approach to gene therapy of gain-of-function receptor mutations.
Collapse
Affiliation(s)
| | - Qiuyan Chen
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232 and
| | - Maria C. Palazzo
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232 and
| | - Evan K. Brooks
- the University of California Los Angeles, Los Angeles, California 90095
| | | | - Tina M. Iverson
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232 and
| | - Wayne L. Hubbell
- the University of California Los Angeles, Los Angeles, California 90095
| | - Vsevolod V. Gurevich
- From the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232 and
| |
Collapse
|
197
|
Ebner K, Muigg P, Singewald N. Inhibitory function of the dorsomedial hypothalamic nucleus on the hypothalamic-pituitary-adrenal axis response to an emotional stressor but not immune challenge. J Neuroendocrinol 2013; 25:48-55. [PMID: 22861486 PMCID: PMC3549560 DOI: 10.1111/j.1365-2826.2012.02369.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 07/24/2012] [Accepted: 07/27/2012] [Indexed: 11/28/2022]
Abstract
Accumulating evidence implicates the dorsomedial hypothalamic nucleus (DMH) in the regulation of autonomic and neuroendocrine stress responses. However, although projections from the DMH to the paraventricular hypothalamic nucleus (PVN), which is the critical site of the neuroendocrine stress axis, have been described, the impact of DMH neurones in the modulation of hypothalamic-pituitary-adrenal (HPA) axis activation during stress is not fully understood. The present study aimed to investigate the role of the DMH in HPA axis responses to different types of stimuli. Male Sprague-Dawley rats fitted with a chronic jugular venous catheter were exposed to either an emotional stressor (elevated platform-exposure) or immune challenge (systemic interleukin-1β administration). Bilateral electrolytic lesions of the DMH disinhibited HPA axis responses to the emotional stressor, as indicated by higher plasma adrenocorticotrophic hormone levels during and after elevated platform exposure in lesioned animals compared to sham-lesioned controls. Moreover, DMH-lesioned animals showed increased neuronal activation in the PVN, as indicated by a higher c-Fos expression after elevated-platform exposure compared to controls. By contrast, DMH-lesions had no effects on HPA axis responses to immune challenge. Taken together, our data suggest an inhibitory role of DMH neurones on stress-induced HPA axis activation that is dependent upon the nature of the stimulus being important in response to an emotional stressor but not to immune challenge.
Collapse
Affiliation(s)
- K Ebner
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens-University of Innsbruck, Innsbruck, Austria.
| | | | | |
Collapse
|
198
|
Gurevich VV, Gurevich EV. Structural determinants of arrestin functions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:57-92. [PMID: 23764050 PMCID: PMC4514030 DOI: 10.1016/b978-0-12-394440-5.00003-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Arrestins are a small protein family with only four members in mammals. Arrestins demonstrate an amazing versatility, interacting with hundreds of different G protein-coupled receptor (GPCR) subtypes, numerous nonreceptor signaling proteins, and components of the internalization machinery, as well as cytoskeletal elements, including regular microtubules and centrosomes. Here, we focus on the structural determinants that mediate various arrestin functions. The receptor-binding elements in arrestins were mapped fairly comprehensively, which set the stage for the construction of mutants targeting particular GPCRs. The elements engaged by other binding partners are only now being elucidated and in most cases we have more questions than answers. Interestingly, even very limited and imprecise identification of structural requirements for the interaction with very few other proteins has enabled the development of signaling-biased arrestin mutants. More comprehensive understanding of the structural underpinning of different arrestin functions will pave the way for the construction of arrestins that can link the receptor we want to the signaling pathway of our choosing.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | | |
Collapse
|
199
|
Walther C, Ferguson SSG. Arrestins: role in the desensitization, sequestration, and vesicular trafficking of G protein-coupled receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:93-113. [PMID: 23764051 DOI: 10.1016/b978-0-12-394440-5.00004-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the years, β-arrestins have emerged as multifunctional molecular scaffolding proteins regulating almost every imaginable G protein-coupled receptor (GPCR) function. Originally discovered as GPCR-desensitizing molecules, they have been shown to also serve as important regulators of GPCR signaling, sequestration, and vesicular trafficking. This broad functional role implicates β-arrestins as key regulatory proteins for cellular function. Hence, this chapter summarizes the current understanding of the β-arrestin family's unique ability to control the kinetics as well as the extent of GPCR activity at the level of desensitization, sequestration, and subsequent intracellular trafficking.
Collapse
Affiliation(s)
- Cornelia Walther
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, Western University Canada, London, Ontario, Canada
| | | |
Collapse
|
200
|
Pascual R, Zamora-León P, Bustamante C. Selegiline (deprenyl) decreases calbindin-D28k expression in cortical neurons of rats socially deprived during the post-weaning period. Int J Dev Neurosci 2012; 31:145-9. [PMID: 23253375 DOI: 10.1016/j.ijdevneu.2012.12.001] [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: 10/19/2012] [Revised: 11/28/2012] [Accepted: 12/05/2012] [Indexed: 11/18/2022] Open
Abstract
Preclinical studies indicate that selegiline (deprenyl), frequently used in some neurodegenerative diseases, exert protective effects on central nervous system neurons of individuals exposed to social isolation (SI). Furthermore, it has been suggested that SI produces neuronal dysfunction due in part to an excessive intracellular Ca(2+) overload. Since the main intracellular Ca(2+) buffering mechanism involves changes in the calcium-binding protein calbindin-D28k (CB), and that CB neuronal expression can increase in response to Ca(2+) transients, we hypothesized that chronic selegiline administration in early socially isolated animals could minimize cell CB expression as an indirect indicator of protective mechanism against Ca(2+) overload. In the present study male rats were weaned at postnatal day 21 (P21) and randomly assigned to social deprivation (SI) or control (SC) environments for 30 days (P21-51). SI animals were further subdivided in two experimental groups: socially deprived-saline (SI-SAL) and socially isolated-selegiline (SI-SEL) for additional 30 days (P52-82). Medial frontal CB immunoreactivity (CB-ir) neurons were quantitatively and qualitatively analyzed. The results obtained indicate that neocortical cells of adult rats submitted to early SI show a significant increase in the number of CB-ir neurons per cortical field, while selegiline treatment significantly reduces this parameter.
Collapse
Affiliation(s)
- Rodrigo Pascual
- Laboratorio de Neurociencias, Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Chile.
| | | | | |
Collapse
|