1
|
Castner SA, Zhang L, Yang CR, Hao J, Cramer JW, Wang X, Bruns RF, Marston H, Svensson KA, Williams GV. Effects of DPTQ, a novel positive allosteric modulator of the dopamine D1 receptor, on spontaneous eye blink rate and spatial working memory in the nonhuman primate. Psychopharmacology (Berl) 2023; 240:1033-1048. [PMID: 36961560 PMCID: PMC10102062 DOI: 10.1007/s00213-022-06282-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/23/2022] [Indexed: 03/25/2023]
Abstract
RATIONALE Dopamine (DA) signaling through the D1 receptor has been shown to be integral to multiple aspects of cognition, including the core process of working memory. The discovery of positive allosteric modulators (PAMs) of the D1 receptor has enabled treatment modalities that may have alternative benefits to orthosteric D1 agonists arising from a synergism of action with functional D1 receptor signaling. OBJECTIVES To investigate this potential, we have studied the effects of the novel D1 PAM DPTQ on a spatial delayed response working memory task in the rhesus monkey. Initial studies indicated that DPTQ binds to primate D1R with high affinity and selectivity and elevates spontaneous eye blink rate in rhesus monkeys in a dose-dependent manner consistent with plasma ligand exposures and central D1activation. RESULTS Based on those results, DPTQ was tested at 2.5 mg/kg IM in the working memory task. No acute effect was observed 1 h after dosing, but performance was impaired 48 h later. Remarkably, this deficit was immediately followed by a significant enhancement in cognition over the next 3 days. In a second experiment in which DPTQ was administered on days 1 and 5, the early impairment was smaller and did not reach statistical significance, but statistically significant enhancement of performance was observed over the following week. Lower doses of 0.1 and 1.0 mg/kg were also capable of producing this protracted enhancement without inducing any transient impairment. CONCLUSIONS DPTQ exemplifies a class of D1PAMs that may be capable of providing long-term improvements in working memory.
Collapse
Affiliation(s)
- Stacy A Castner
- Department of Comparative Medicine, Yale University, 310 Cedar St, New Haven, CT, 06520, USA
| | - Linli Zhang
- ChemPartner, 99 Lian He North Road, Zhe Lin Town, Fengxian Area, Shanghai, China
| | - Charles R Yang
- ChemPartner, 99 Lian He North Road, Zhe Lin Town, Fengxian Area, Shanghai, China
| | - Junliang Hao
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Jeffrey W Cramer
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Xushan Wang
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Robert F Bruns
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | | | - Kjell A Svensson
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Graham V Williams
- Department of Comparative Medicine, Yale University, 310 Cedar St, New Haven, CT, 06520, USA.
| |
Collapse
|
2
|
Santos PI, Outeiro TF. Protein trapping leads to altered synaptic proteostasis in synucleinopathies. FEBS J 2020; 287:5294-5303. [PMID: 32400966 DOI: 10.1111/febs.15364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/01/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is associated with the accumulation of alpha-synuclein (aSyn) in intracellular inclusions known as Lewy bodies and Lewy neurites. Under physiological conditions, aSyn is found at the presynaptic terminal and exists in a dynamic equilibrium between soluble, membrane-associated and aggregated forms. Emerging evidence suggests that, under pathological conditions, aSyn begins to accumulate and acquire a toxic function at the synapse, impairing their normal function and connectivity. However, the precise molecular mechanisms linking aSyn accumulation and synaptic dysfunction are still elusive. Here, we provide an overview of our current findings and discuss the hypothesis that certain aSyn aggregates may interact with proteins with whom aSyn normally does not interact with, thereby trapping them and preventing them from performing their normal functions in the cell. We posit that such abnormal interactions start to occur during the prodromal stages of PD, eventually resulting in the overt manifestation of clinical features. Therefore, understanding the nature and behaviour of toxic aSyn species and their contribution to aSyn-mediated toxicity is crucial for the development of therapeutic strategies capable of modifying disease progression in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Patrícia I Santos
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
3
|
Park HJ, Zhao TT, Park KH, Lee MK. Repeated treatments with the D 1 dopamine receptor agonist SKF-38393 modulate cell viability via sustained ERK-Bad-Bax activation in dopaminergic neuronal cells. Behav Brain Res 2019; 367:166-175. [PMID: 30930179 DOI: 10.1016/j.bbr.2019.03.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/21/2019] [Accepted: 03/19/2019] [Indexed: 01/23/2023]
Abstract
The D1 dopamine receptor agonist, SKF-38393, induces cytotoxicity in striatal dopaminergic neurons via an extracellular signal-regulated kinase (ERK) signaling cascade. However, the underlying mechanism remains unclear. We hypothesized that repeated activation of dopaminergic receptors by agonists could lead to neuronal cell death. This study investigated the effects of SKF-38393 on dopaminergic neuronal cell death in a 6-hydroxydopamine-lesioned rat model of Parkinson's disease (PD) and PC12 cells. In the PD model, SKF-38393 administration (3 and 10 mg/kg per day, s.c.) for 8 weeks significantly increased the number of tyrosine hydroxylase-immunopositive neuronal cells in nigrostriatal regions. SKF-38393 administration for 8 weeks induced phosphorylation of sustained ERK1/2 and Bad (Bcl-2-associated death promoter) at Ser155 (BadSer155), and augmented Bax (Bcl-2-associated X protein) expression. However, SKF-38393 only increased Bad phosphorylation at Ser112 (BadSer112) when administered for 4 weeks. In PC12 cells, toxic levels of SKF-38393 (20 and 50 μM) rapidly induced formation of neurite-like processes, but not in the presence of an adenylyl cyclase inhibitor (MDL-12330 A). SKF-38393 (20 and 50 μM) induced sustained ERK1/2 and BadSer155 phosphorylation as well as caspase-3 activation. At a non-toxic level (5 μM), SKF-38393 produced only transient ERK1/2 and BadSer112 phosphorylation. Repeated treatments with SKF-38393 (5 μM) for 1-3 days activated BadSer112. Repeated treatments for 4-7 days induced sustained ERK1/2 and BadSer155 phosphorylation as well as Bax and caspase-3 activation. These results suggest that SKF-38393 induces neurotoxicity by activation of the sustained ERK-Bad-Bax system. These findings contribute to an understanding of the adverse effects of D1 dopamine receptor agonists in patients with PD.
Collapse
Affiliation(s)
- Hyun Jin Park
- Department of Pharmacy and Research Center for Bioresource and Health, College of Pharmacy, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Ting Ting Zhao
- Department of Pharmacy and Research Center for Bioresource and Health, College of Pharmacy, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Keun Hong Park
- Department of Pharmacy and Research Center for Bioresource and Health, College of Pharmacy, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong, Heungduk-gu, Cheongju 28160, Republic of Korea
| | - Myung Koo Lee
- Department of Pharmacy and Research Center for Bioresource and Health, College of Pharmacy, Chungbuk National University, 194-21, Osongsaengmyeong 1-ro, Osong, Heungduk-gu, Cheongju 28160, Republic of Korea.
| |
Collapse
|
4
|
Helbing C, Tischmeyer W, Angenstein F. Late effect of dopamine D 1/5 receptor activation on stimulus-induced BOLD responses in the hippocampus and its target regions depends on the history of previous stimulations. Neuroimage 2017; 152:119-129. [DOI: 10.1016/j.neuroimage.2017.02.077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/23/2017] [Accepted: 02/25/2017] [Indexed: 10/20/2022] Open
|
5
|
Li Q, Wu N, Cui P, Gao F, Qian WJ, Miao Y, Sun XH, Wang Z. Suppression of outward K(+) currents by activating dopamine D1 receptors in rat retinal ganglion cells through PKA and CaMKII signaling pathways. Brain Res 2016; 1635:95-104. [PMID: 26826585 DOI: 10.1016/j.brainres.2016.01.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/17/2016] [Accepted: 01/21/2016] [Indexed: 01/11/2023]
Abstract
Dopamine plays an important role in regulating neuronal functions in the central nervous system by activating the specific G-protein coupled receptors. Both D1 and D2 dopamine receptors are extensively distributed in the retinal neurons. In the present study, we investigated the effects of D1 receptor signaling on outward K(+) currents in acutely isolated rat retinal ganglion cells (RGCs) by patch-clamp techniques. Extracellular application of SKF81297 (10 μM), a specific D1 receptor agonist, significantly and reversibly suppressed outward K(+) currents of the cells, which was reversed by SCH23390 (10 μM), a selective D1 receptor antagonist. We further showed that SKF81297 mainly suppressed the glybenclamide (Gb)- and 4-aminopyridine (4-AP)-sensitive K(+) current components, but did not show effect on the tetraethylammonium (TEA)-sensitive one. Both protein kinase A (PKA) and calcium/calmodulin-dependent protein kinase II (CaMKII) signaling pathways were likely involved in the SKF81297-induced suppression of the K(+) currents since either Rp-cAMP (10 μM), a cAMP/PKA signaling inhibitor, or KN-93 (10 μM), a specific CaMKII inhibitor, eliminated the SKF81297 effect. In contrast, neither protein kinase C (PKC) nor mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway seemed likely to be involved because both the PKC inhibitor bisindolylmaleimide IV (Bis IV) (10 μM) and the MAPK/ERK1/2 inhibitor U0126 (10 μM) did not block the SKF81297-induced suppression of the K(+) currents. These results suggest that activation of D1 receptors suppresses the Gb- and 4-AP-sensitive K(+) current components in rat RGCs through the intracellular PKA and CaMKII signaling pathways, thus modulating the RGC excitability.
Collapse
Affiliation(s)
- Qian Li
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Na Wu
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Peng Cui
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Feng Gao
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Wen-Jing Qian
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Yanying Miao
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Xing-Huai Sun
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Zhongfeng Wang
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
6
|
Shivarama Shetty M, Gopinadhan S, Sajikumar S. Dopamine D1/D5 receptor signaling regulates synaptic cooperation and competition in hippocampal CA1 pyramidal neurons via sustained ERK1/2 activation. Hippocampus 2015; 26:137-50. [PMID: 26194339 PMCID: PMC5054950 DOI: 10.1002/hipo.22497] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2015] [Indexed: 12/30/2022]
Abstract
Synaptic cooperation and competition are important components of synaptic plasticity that tune synapses for the formation of associative long‐term plasticity, a cellular correlate of associative long‐term memory. We have recently reported that coincidental activation of weak synapses within the vicinity of potentiated synapses will alter the cooperative state of synapses to a competitive state thus leading to the slow decay of long‐term plasticity, but the molecular mechanism underlying this is still unknown. Here, using acute hippocampal slices of rats, we have examined how increasing extracellular dopamine concentrations interact and/or affect electrically induced long‐term potentiation (LTP) in the neighboring synapses. We demonstrate that D1/D5‐receptor‐mediated potentiation at the CA1 Schaffer collateral synapses differentially regulates synaptic co‐operation and competition. Further investigating the molecular players involved, we reveal an important role for extracellular signal‐regulated kinases‐1 and 2 (ERK1/2) as signal integrators and dose‐sensors. Interestingly, a sustained activation of ERK1/2 pathway seems to be involved in the differential regulation of synaptic associativity. The concentration‐dependent effects of the modulatory transmitter, as demonstrated for dopaminergic signaling in the present study, might offer additional computational power by fine tuning synaptic associativity processes for establishing long‐term associative memory in neural networks. © 2015 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mahesh Shivarama Shetty
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, Singapore
| | - Suma Gopinadhan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, Singapore
| |
Collapse
|
7
|
Bhatt DK, Ramachandran R, Christensen SLT, Gupta S, Jansen-Olesen I, Olesen J. CGRP infusion in unanesthetized rats increases expression of c-Fos in the nucleus tractus solitarius and caudal ventrolateral medulla, but not in the trigeminal nucleus caudalis. Cephalalgia 2014; 35:220-33. [PMID: 24895375 DOI: 10.1177/0333102414535995] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND AIMS Calcitonin gene-related peptide (CGRP) and glyceryl trinitrate (GTN) infusion in migraineurs provokes headache resembling spontaneous migraine, and CGRP receptor antagonists are effective in the treatment of acute migraine. We hypothesized that CGRP infusion would increase molecular markers of neuronal activation in migraine-relevant tissues of the rat. METHODS CGRP was infused intravenously (i.v.) in freely moving rats to circumvent factors like anesthesia, acute surgery and severe hypotension, the three confounding factors for c-Fos expression. The trigeminal nucleus caudalis (TNC) was isolated at different time points after CGRP infusion. The level of c-Fos mRNA and protein expression in TNC were analyzed by qPCR and immunohistochemistry. c-Fos-stained nuclei were also counted in the nucleus tractus solitarius (NTS) and caudal ventrolateral medulla (CVLM), integrative sites in the brain stem for processing cardiovascular signals. We also investigated Zif268 protein expression (another immediate early gene) in TNC. The protein expression of p-ERK, p-CREB and c-Fos was analyzed in dura mater, trigeminal ganglion (TG) and TNC samples using Western blot. RESULTS CGRP infusion caused a significant dose-dependent fall in mean arterial blood pressure. No significant activation of c-Fos in the TNC at mRNA and protein levels was observed after CGRP infusion. A significant increase in c-Fos protein was observed in the NTS and CVLM in the brain stem. Zif268 expression in the TNC was also not changed after CGRP infusion. p-ERK was increased in the dura mater 30 minutes after CGRP infusion. CONCLUSION CGRP infusion increased the early expression of p-ERK in the dura mater but did not increase c-Fos and Zif268 expression in the TNC. The rats may, thus, differ from migraine patients, in whom infusion of CGRP caused headache and a delayed migraine attack. The rat CGRP infusion model with c-Fos or Zif268 as neuronal pain markers in TNC is unsuitable for antimigraine drug testing.
Collapse
Affiliation(s)
| | | | | | | | | | - Jes Olesen
- Department of Neurology, Glostrup Hospital, Denmark
| |
Collapse
|
8
|
Zhong SY, Chen YX, Fang M, Zhu XL, Zhao YX, Liu XY. Low-dose levodopa protects nerve cells from oxidative stress and up-regulates expression of pCREB and CD39. PLoS One 2014; 9:e95387. [PMID: 24743653 PMCID: PMC3990701 DOI: 10.1371/journal.pone.0095387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 03/25/2014] [Indexed: 11/18/2022] Open
Abstract
Objective This study aimed to investigate the influence of low-dose levodopa (L-DOPA) on neuronal cell death under oxidative stress. Methods PC12 cells were treated with L-DOPA at different concentrations. We detected the L-DOPA induced reactive oxygen species (ROS). Meanwhile, MTT and LDH assay were performed to determine the proliferation and growth of PC12 cells with or without ROS scavenger. In addition, after pretreatment with L-DOPA at different concentrations alone or in combination with CD39 inhibitor, PC12 cells were incubated with hydrogen peroxide (H2O2) and the cell viability was evaluated by MTT and LDH assay. In addition, the expression of pCREB and CD39 was detected by immunofluorescence staining and Western blot assay in both cells and rat’s brain after L-DOPA treatment. Results After treatment with L-DOPA for 3 days, the cell proliferation and growth were promoted when the L-DOPA concentration was <30 µM, while cell proliferation was comparable to that in control group when the L-DOPA concentration was >30 µM. Low dose L-DOPA could protect the PC12 cells from H2O2 induced oxidative stress, which was compromised by CD39 inhibitor. In addition, the expression of CD39 and pCREB increased in both PC12 cells and rats’ brain after L-DOPA treatment. Conclusions L-DOPA at different concentrations has distinct influence on proliferation and growth of PC12 cells, and low dose (<30 µM) L-DOPA protects PC12 cells against oxidative stress which might be related to the up-regulation of CD39 and pCREB expression.
Collapse
Affiliation(s)
- Shi-Ying Zhong
- Department of Neurology, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Shanghai, China
| | - Yong-Xing Chen
- Department of Neurology, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Shanghai, China
| | - Min Fang
- Department of Neurology, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Shanghai, China
| | - Xiao-Long Zhu
- Department of Neurology, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Shanghai, China
| | - Yan-Xin Zhao
- Department of Neurology, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Shanghai, China
- * E-mail: (YXZ); (XYL)
| | - Xue-Yuan Liu
- Department of Neurology, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Shanghai, China
- * E-mail: (YXZ); (XYL)
| |
Collapse
|
9
|
Lindenbach D, Dupre KB, Eskow Jaunarajs KL, Ostock CY, Goldenberg AA, Bishop C. Effects of 5-HT1A receptor stimulation on striatal and cortical M1 pERK induction by L-DOPA and a D1 receptor agonist in a rat model of Parkinson's disease. Brain Res 2013; 1537:327-39. [PMID: 24060645 DOI: 10.1016/j.brainres.2013.09.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 09/09/2013] [Accepted: 09/17/2013] [Indexed: 11/17/2022]
Abstract
Motor symptoms of Parkinson's disease are commonly treated using l-DOPA although long-term treatment usually causes debilitating motor side effects including dyskinesias. A putative source of dyskinesia is abnormally high levels of phosphorylated extracellular-regulated kinase (pERK) within the striatum. In animal models, the serotonin 1A receptor agonist ±8-OH-DPAT reduces dyskinesia, suggesting it may exhibit efficacy through the pERK pathway. The present study investigated the effects of ±8-OH-DPAT on pERK density in rats treated with l-DOPA or the D1 receptor agonist SKF81297. Rats were given a unilateral dopamine lesion with 6-hydroxydopamine and primed with a chronic regimen of l-DOPA, SKF81297 or their vehicles. On the final test day, rats were given two injections: first with ±8-OH-DPAT, the D1 receptor antagonist SCH23390 or their vehicles, and second with l-DOPA, SKF81297 or their vehicles. Rats were then transcardially perfused for immunohistological analysis of pERK expression in the striatum and primary motor cortex. Rats showed greater dyskinesia in response to l-DOPA and SKF81297 after repeated injections. Although striatal pERK induction was similar between acute and chronic l-DOPA, SKF81297 caused the largest increase in striatal pERK after the first exposure. Neither compound alone affected motor cortex pERK. Surprisingly, in the ventromedial striatum, ±8-OH-DPAT potentiated l-DOPA-induced pERK; in the motor cortex, ±8-OH-DPAT potentiated pERK with l-DOPA or SKF81297. Our results support previous work that the striatal pERK pathway is dysregulated after dopamine depletion, but call into question the utility of pERK as a biomarker of dyskinesia expression.
Collapse
Affiliation(s)
- David Lindenbach
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University - State University of New York, Binghamton, NY, USA
| | | | | | | | | | | |
Collapse
|
10
|
Zhang L, Chen QP, Wang JK, Zhang LC, Zeng YM. Effects of morphine-dependent and withdrawal on activation of the distal cerebrospinal fluid contacting neurons' phosphorylation CREB in rat brain. Neurol Res 2013; 31:738-42. [DOI: 10.1179/174313209x382386] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
11
|
Wang M, Wong AH, Liu F. Interactions between NMDA and dopamine receptors: A potential therapeutic target. Brain Res 2012; 1476:154-63. [DOI: 10.1016/j.brainres.2012.03.029] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 03/08/2012] [Accepted: 03/10/2012] [Indexed: 11/26/2022]
|
12
|
Hu Q, Li B, Xu R, Chen D, Mu C, Fei E, Wang G. The protease Omi cleaves the mitogen-activated protein kinase kinase MEK1 to inhibit microglial activation. Sci Signal 2012; 5:ra61. [PMID: 22912494 DOI: 10.1126/scisignal.2002946] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Inflammation in Parkinson's disease is closely associated with disease pathogenesis. Mutations in Omi, which encodes the protease Omi, are linked to neurodegeneration and Parkinson's disease in humans and in mouse models. The severe neurodegeneration and neuroinflammation that occur in mnd2 (motor neuron degeneration 2) mice result from loss of the protease activity of Omi by the point mutation S276C; however, the substrates of Omi that induce neurodegeneration are unknown. We showed that Omi was required for the production of inflammatory molecules by microglia, which are the resident macrophages in the central nervous system. Omi suppressed the activation of the mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase 1 and 2 (ERK1/2) by cleaving the upstream kinase MEK1 (mitogen-activated or extracellular signal-regulated protein kinase kinase 1). Knockdown of Omi in microglial cell lines led to activation of ERK1/2 and resulted in degradation of IκBα [α inhibitor of nuclear factor κB (NF-κB)], resulting in NF-κB activation and the expression of genes encoding inflammatory molecules, such as tumor necrosis factor-α and inducible nitric oxide synthase. The production of inflammatory molecules induced by the knockdown of Omi was blocked by the MEK1-specific inhibitor U0126. Furthermore, expression of the protease-deficient S276C Omi mutant in a microglial cell line had no effect on MEK1 cleavage or ERK1/2 activation. In the brains of mnd2 mice, we observed increased transcription of several genes encoding inflammatory molecules, as well as activation of astrocytes and microglia. Therefore, our study demonstrates that Omi is an intrinsic cellular factor that inhibits neuroinflammation.
Collapse
Affiliation(s)
- Qingsong Hu
- Laboratory of Molecular Neuropathology, Key Laboratory of Brain Function and Diseases and School of Life Sciences, University of Science and Technology of China, Chinese Academy of Sciences, Hefei, Anhui 230027, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Gangarossa G, Valjent E. Regulation of the ERK pathway in the dentate gyrus by in vivo dopamine D1 receptor stimulation requires glutamatergic transmission. Neuropharmacology 2012; 63:1107-17. [PMID: 22796106 DOI: 10.1016/j.neuropharm.2012.06.062] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 06/22/2012] [Accepted: 06/29/2012] [Indexed: 11/18/2022]
Abstract
Acute systemic administration of the dopamine D1/D5 receptors (D1Rs) agonist, SKF81297, activates the extracellular signal-regulated protein kinases (ERK) pathway selectively in the granule cells of the dentate gyrus. In this study, we examined the mechanisms involved in this regulation and investigated the molecular components that could promote ERK-dependent transcription and translation. SKF81297 induced phosphorylation of ERK and histone H3 required intact glutamatergic transmission. Blockade of glutamate release achieved by the mGluR2/3 agonist, LY354740 or the selective adenosine A1R agonist, CCPA as well as neurotoxic lesions of lateral entorhinal cortex reduced the ability of SKF81297 to induce ERK activation in the dentate gyrus. This activation required the combined stimulation of NR2B-containing NMDARs, mGluR1 and mGluR5. SKF81297 evoked phosphorylation of the ribosomal protein S6 (rpS6) selectively at the Ser235/236 site while the Ser240/244 site remains unchanged. The SKF81297 induced increased phosphorylation of rpS6 was dependent on PKC and ERK/p90RSK activation. Surprisingly, administration of D1Rs agonist suppressed mTORC1/p70S6K pathway suggesting an mTOR-independent regulation of rpS6 phosphorylation. Taken together, our results show that intact glutamatergic transmission plays a major role in the regulation of ERK-dependent phosphorylation of histone H3 and rpS6 observed in the mouse dentate gyrus after systemic administration of SKF81297.
Collapse
|
14
|
Moreno C, Sivam SP. The Time Course of D1 Agonist Induced Striatonigral ERK1/2 Signaling in a Rat Model of Parkinson’s Disease. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/jbbs.2012.21001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
15
|
Gafford GM, Parsons RG, Helmstetter FJ. Consolidation and reconsolidation of contextual fear memory requires mammalian target of rapamycin-dependent translation in the dorsal hippocampus. Neuroscience 2011; 182:98-104. [PMID: 21439355 PMCID: PMC3087706 DOI: 10.1016/j.neuroscience.2011.03.023] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 03/02/2011] [Accepted: 03/09/2011] [Indexed: 12/26/2022]
Abstract
The mammalian target of rapamycin (mTOR) pathway is important for regulating protein translation. The present study characterized the role of mTOR-dependent translation in the dorsal hippocampus (DH) during the consolidation and reconsolidation of contextual fear memory. We first showed that fear conditioning resulted in increased phosphorylation of p70s6 kinase (p70s6K) in the DH and that infusion of the mTOR inhibitor rapamycin (RAP) into the DH immediately after training disrupted formation of long-term contextual fear memory. Additionally we showed that p70s6K was activated after retrieval of a previously stored fear memory, and inhibition of mTOR by DH infusion of RAP blocked the reconsolidation of contextual fear memory. Together these results demonstrate that within the DH translational control through the mTOR pathway is important for consolidation as well as the stability of fear memory after retrieval.
Collapse
Affiliation(s)
- G M Gafford
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA.
| | | | | |
Collapse
|
16
|
Papadeas ST, Halloran C, McCown TJ, Breese GR, Blake BL. Changes in apical dendritic structure correlate with sustained ERK1/2 phosphorylation in medial prefrontal cortex of a rat model of dopamine D1 receptor agonist sensitization. J Comp Neurol 2008; 511:271-85. [PMID: 18785628 PMCID: PMC2587500 DOI: 10.1002/cne.21835] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Rats lesioned with 6-hydroxydopamine (6-OHDA) as neonates exhibit behavioral and neurochemical abnormalities in adulthood that mimic Lesch-Nyhan disease, schizophrenia, and other developmental disorders of frontostriatal circuit dysfunction. In these animals a latent sensitivity to D1 agonists is maximally exposed by repeated administration of dopamine agonists in the postpubertal period (D1 priming). In neonate-lesioned, adult rats primed with SKF-38393, we found selective, persistent alterations in the morphology of pyramidal neuron apical dendrites in the prelimbic area of the medial prefrontal cortex (mPFC). In these animals, dendrite bundling patterns and the typically straight trajectories of primary dendritic shafts were disrupted, whereas the diameter of higher-order oblique branches was increased. Although not present in neonate-lesioned rats treated with saline, these morphological changes persisted at least 21 days after repeated dosing with SKF-38393, and were not accompanied by markers of neurodegenerative change. A sustained increase in phospho-ERK immunoreactivity in wavy dendritic shafts over the same period suggested a relationship between prolonged ERK phosphorylation and dendritic remodeling in D1-primed rats. In support of this hypothesis, pretreatment with the MEK1/2-ERK1/2 pathway inhibitors PD98059 or SL327, prior to each priming dose of SKF-38393, prevented the morphological changes associated with D1 priming. Together, these findings demonstrate that repeated stimulation of D1 receptors in adulthood interacts with the developmental loss of dopamine to profoundly and persistently modify neuronal signaling and dendrite morphology in the mature prefrontal cortex. Furthermore, sustained elevation of ERK activity in mPFC pyramidal neurons may play a role in guiding these morphological changes in vivo.
Collapse
Affiliation(s)
- Sophia T. Papadeas
- GRADUATE PROGRAM IN NEUROBIOLOGY, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
- BOWLES CENTER FOR ALCOHOL STUDIES, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
| | - Christopher Halloran
- BOWLES CENTER FOR ALCOHOL STUDIES, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
| | - Thomas J. McCown
- BOWLES CENTER FOR ALCOHOL STUDIES, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
- GENE THERAPY CENTER, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
- DEPARTMENT OF PSYCHIATRY, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
| | - George R. Breese
- GRADUATE PROGRAM IN NEUROBIOLOGY, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
- BOWLES CENTER FOR ALCOHOL STUDIES, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
- DEPARTMENT OF PSYCHIATRY, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
- DEPARTMENT OF PHARMACOLOGY, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
| | - Bonita L. Blake
- BOWLES CENTER FOR ALCOHOL STUDIES, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
- DEPARTMENT OF PSYCHIATRY, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
- DEPARTMENT OF PHARMACOLOGY, SCHOOL OF MEDICINE, UNIVERSITY OF NORTH CAROLINA at CHAPEL HILL, CHAPEL HILL, NC 27599
| |
Collapse
|
17
|
Schicknick H, Schott BH, Budinger E, Smalla KH, Riedel A, Seidenbecher CI, Scheich H, Gundelfinger ED, Tischmeyer W. Dopaminergic modulation of auditory cortex-dependent memory consolidation through mTOR. ACTA ACUST UNITED AC 2008; 18:2646-58. [PMID: 18321872 PMCID: PMC2567422 DOI: 10.1093/cercor/bhn026] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Previous studies in the auditory cortex of Mongolian gerbils on discrimination learning of the direction of frequency-modulated tones (FMs) revealed that long-term memory formation involves activation of the dopaminergic system, activity of the protein kinase mammalian target of rapamycin (mTOR), and protein synthesis. This led to the hypothesis that the dopaminergic system might modulate memory formation via regulation of mTOR, which is implicated in translational control. Here, we report that the D1/D5 dopamine receptor agonist SKF-38393 substantially improved gerbils’ FM discrimination learning when administered systemically or locally into the auditory cortex shortly before, shortly after, or 1 day before conditioning. Although acquisition performance during initial training was normal, the discrimination of FMs was enhanced during retraining performed hours or days after agonist injection compared with vehicle-injected controls. The D1/D5 receptor antagonist SCH-23390, the mTOR inhibitor rapamycin, and the protein synthesis blocker anisomycin suppressed this effect. By immunohistochemistry, D1 dopamine receptors were identified in the gerbil auditory cortex predominantly in the infragranular layers. Together, these findings suggest that in the gerbil auditory cortex dopaminergic inputs regulate mTOR-mediated, protein synthesis-dependent mechanisms, thus controlling for hours or days the consolidation of memory required for the discrimination of complex auditory stimuli.
Collapse
|
18
|
Sivam SP, Pugazhenthi S, Pugazhenthi V, Brown H. L-DOPA-induced activation of striatal p38MAPK and CREB in neonatal dopaminergic denervated rat: Relevance to self-injurious behavior. J Neurosci Res 2008; 86:339-49. [PMID: 17893915 DOI: 10.1002/jnr.21504] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The destruction of nigrostriatal dopaminergic neurons with 6-hydroxydopamine (6OHDA) during the neonatal period results in dopamine (DA) loss and susceptibility for self-injurious behavior (SIB) when challenged with L-dihydroxyphenylalanine (L-DOPA), via a supersensitive D1 receptor-mediated mechanism. However, there are no changes in D1 receptor binding or mRNA levels, suggesting a potential postreceptor signaling mechanism(s). Here, we examined whether L-DOPA-induced SIB is associated with altered MAPK signaling (p38MAPK, ERK1/2, and JNK) and their nuclear target, CREB. Neonatal dopaminergic lesioned animals were challenged, as adults, with L-DOPA, observed for SIB for 6 hr, and then sacrificed. The data were grouped as follows: control, lesioned rats without SIB (SIB(-)), and lesioned rats that were positive for SIB (SIB(+)). HPLC analysis of striatal extracts revealed a more significant loss of DA and an increase of serotonin in the SIB(+) than in the SIB(-) group. The striatal levels of TH protein were severely decreased, but D1 receptor levels were unaltered in the lesioned groups. These results confirm and extend previous studies indicating that SIB is associated with a near-total loss of DA and TH, an increase in serotonin, and no change in D1 receptor levels. The present studies further revealed that the levels of active phosphorylated forms of p38MAPK and CREB were significantly higher in the SIB(+) group than in the SIB(-) group in the striatum, but not in cortex or olfactory tubercle. The results indicate an induction of striatal p38MAPK and an activation of its nuclear target, CREB, as additional mechanisms in the genesis of L-DOPA-induced SIB.
Collapse
Affiliation(s)
- Subbiah P Sivam
- Department of Pharmacology and Toxicology, Indiana University School of Medicine-Northwest, Gary, IN 46408, USA.
| | | | | | | |
Collapse
|
19
|
Chu CT, Plowey ED, Wang Y, Patel V, Jordan-Sciutto KL. Location, location, location: altered transcription factor trafficking in neurodegeneration. J Neuropathol Exp Neurol 2007; 66:873-83. [PMID: 17917581 PMCID: PMC2220049 DOI: 10.1097/nen.0b013e318156a3d7] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neurons may be particularly sensitive to disruptions in transcription factor trafficking. Survival and injury signals must traverse dendrites or axons, in addition to soma, to affect nuclear transcriptional responses. Transcription factors exhibit continued nucleocytoplasmic shuttling; the predominant localization is regulated by binding to anchoring proteins that mask nuclear localization/export signals and/or target the factor for degradation. Two functional groups of karyopherins, importins and exportins, mediate RanGTPase-dependent transport through the nuclear pore. A growing number of recent studies, in Alzheimer, Parkinson, and Lewy body diseases, amyotrophic lateral sclerosis, and human immunodeficiency virus encephalitis, implicate aberrant cytoplasmic localization of transcription factors and their regulatory kinases in degenerating neurons. Potential mechanisms include impaired nuclear import, enhanced export, suppression of degradation, and sequestration in protein aggregates or organelles and may reflect unmasking of alternative cytoplasmic functions, both physiologic and pathologic. Some "nuclear" factors also function in mitochondria, and importins are also involved in axonal protein trafficking. Detrimental consequences of a decreased nuclear to cytoplasmic balance include suppression of neuroprotective transcription mediated by cAMP- and electrophile/antioxidant-response elements and gain of toxic cytoplasmic effects. Studying the pathophysiologic mechanisms regulating transcription factor localization should facilitate strategies to bypass deficits and restore adaptive neuroprotective transcriptional responses.
Collapse
Affiliation(s)
- Charleen T Chu
- Department of Pathology, Division of Neuropathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | |
Collapse
|
20
|
Abstract
The neurotoxin 6-hydroxydopamine (6-OHDA) continues to constitute a valuable topical tool used chiefly in modeling Parkinson's disease in the rat. The classical method of intracerebral infusion of 6-OHDA involving a massive destruction of nigrostriatal dopaminergic neurons, is largely used to investigate motor and biochemical dysfunctions in Parkinson's disease. Subsequently, more subtle models of partial dopaminergic degeneration have been developed with the aim of revealing finer motor deficits. The present review will examine the main features of 6-OHDA models, namely the mechanisms of neurotoxin-induced neurodegeneration as well as several behavioural deficits and motor dysfunctions, including the priming model, modeled by this means. An overview of the most recent morphological and biochemical findings obtained with the 6-OHDA model will also be provided, particular attention being focused on the newly investigated intracellular mechanisms at the striatal level (e.g., A(2A) and NMDA receptors, PKA, CaMKII, ERK kinases, as well as immediate early genes, GAD67 and peptides). Thanks to studies performed in the 6-OHDA model, all these mechanisms have now been hypothesised to represent the site of pathological dysfunction at cellular level in Parkinson's disease.
Collapse
Affiliation(s)
- Nicola Simola
- Department of Toxicology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | | | | |
Collapse
|
21
|
Beard JL, Unger EL, Bianco LE, Paul T, Rundle SE, Jones BC. Early postnatal iron repletion overcomes lasting effects of gestational iron deficiency in rats. J Nutr 2007; 137:1176-82. [PMID: 17449578 DOI: 10.1093/jn/137.5.1176] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Iron deficiency anemia in early childhood causes developmental delays and, very likely, irreversible alterations in neurological functioning. One primary goal for the present study was to determine whether the effects of late gestational iron deficiency on brain monoamine metabolism, iron content, and behavioral phenotypes could be repaired with iron intervention in early lactation. Young pregnant rats were provided iron-deficient or control diets from mid-gestation (G15). At postnatal d 4 (P4), pups from iron-deficient dams were out-fostered either to other ID dams or control dams while pups of control dams were similarly fostered to other control dams. Dietary treatments continued to adulthood (P65) when brain iron and regional monoamines were evaluated. P4 iron repletion normalized body iron status, brain iron concentrations, monoamine concentrations, and monoamine transporter and receptor densities in most brain regions. Dopamine transporter densities in caudate and substantia nigra were lower in ID rats but were normalized with iron repletion. Serotonin transporter levels in most brain regions and open-field exploration were also normalized with iron repletion. The success of this approach of early postnatal iron intervention following iron deficiency in utero contrasts to a relative lack of success when the intervention is performed at weaning. These data suggest that a window of opportunity exists for reversing the detrimental effects of iron deficiency in utero in rats and provides strong support of intervention approaches in humans with iron deficiency during pregnancy.
Collapse
Affiliation(s)
- John L Beard
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Milasin JM, Buffo A, Carulli D, Strata P. Intensive remodeling of Purkinje cell spines after climbing fibers deafferentation does not involve MAPK and Akt activation. Ann N Y Acad Sci 2007; 1096:230-8. [PMID: 17405934 DOI: 10.1196/annals.1397.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Subtotal lesion of the inferior olive (IO) achieved by treating experimental animals with 3-acetylpyridine (3AP) induces partial Purkinje cells (PCs) deafferentation that leads to PC hyperactivity and new spine formation. Coincidentally, the olivary terminals belonging to the few survived olivary neurons undergo an extensive collateral sprouting resulting in reinnervation of the neighboring denervated PCs. We obtained chemical deafferentation of PCs in adult rats (body weight, 120-170 g; age, 35-40 days) by a single intraperitoneal injection of 3AP (65 mg/kg body weight), and as early as 3 days after 3AP treatment, important morphological changes could be observed on PCs. Mitogen-activated protein kinase (MAPK) cascades and more specifically extracellular signal-regulated kinases 1/2 (ERK1/2) play a critical role in the signaling events underlying synaptic plasticity. For instance, long-term depression (LTD) in the adult hippocampus and long-term potentiation (LTP) in cerebellum both involve ERK activation. Since PCs deprived of their climbing fibers (CFs) afferents initiate an intensive remodeling of the spines and rapid recall of the remaining CFs, it prompted us to see whether the observed phenomena correlated with MAPK and Akt activation. Immunohistochemistry and Western blotting were done at various time points after 3AP application (from 24 h to 6 days), as the exact dynamics of CF loss is not precisely known. As judged by Western blotting, there was no increase of activated ERK in the cerebellum. However, immunohistochemistry revealed increased ERK phosphorylation in the "pinceaux" of basket cells in 3AP animals. Similarly, stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK), p38 MAPK, and Akt activation were also studied by means of Western blotting and immunohistochemistry. Upon 3AP treatment no changes in phosphorylation status could be seen in the different kinases subjected to analysis. Our results suggest that activation of MAPK and Akt cascades is not essential in this model of neuronal plasticity.
Collapse
Affiliation(s)
- Jelena M Milasin
- Institute of Biology and Human Genetics, School of Dentistry, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia and Montenegro.
| | | | | | | |
Collapse
|
23
|
Castner SA, Williams GV. Tuning the engine of cognition: A focus on NMDA/D1 receptor interactions in prefrontal cortex. Brain Cogn 2007; 63:94-122. [PMID: 17204357 DOI: 10.1016/j.bandc.2006.11.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 11/05/2006] [Accepted: 11/08/2006] [Indexed: 11/18/2022]
Abstract
The prefrontal cortex of the primate frontal lobes provides the capacity for judgment which can constantly adapt behavior in order to optimize its outcome. Adjudicating between long-term memory programs and prepotent responses, this capacity reviews all incoming information and provides an interpretation dependent on the events that have just occurred, the events that are predicted to happen, and the alternative response strategies that are available in the given situation. It has been theorized that this function requires two essential integrated components, a central executive which guides selective attention based on mechanisms of associative memory, as well as the second component, working memory buffers, in which information is held online, abstracted, and translated on a mental sketchpad of work in progress. In this review, we critically outline the evidence that the integration of these processes and, in particular, the induction and maintenance of persistent activity in prefrontal cortex and related networks, is dependent upon the interaction of dopamine D1 and glutamate NMDA receptor signaling at critical nodes within local circuits and distributed networks. We argue that this interaction is not only essential for representational memory, but also core to mechanisms of neuroadaptation and learning. Understanding its functional significance promises to reveal major new insights into prefrontal dysfunction in schizophrenia and, hence, to target a new generation of drugs designed to ameliorate the debilitating working memory deficits in this disorder.
Collapse
Affiliation(s)
- Stacy A Castner
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA.
| | | |
Collapse
|
24
|
Repunte-Canonigo V, Lutjens R, van der Stap LD, Sanna PP. Increased expression of protein kinase A inhibitor alpha (PKI-alpha) and decreased PKA-regulated genes in chronic intermittent alcohol exposure. Brain Res 2007; 1138:48-56. [PMID: 17270154 PMCID: PMC4485929 DOI: 10.1016/j.brainres.2006.09.115] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2006] [Revised: 09/19/2006] [Accepted: 09/29/2006] [Indexed: 11/17/2022]
Abstract
Intermittent models of alcohol exposure that mimic human patterns of alcohol consumption produce profound physiological and biochemical changes and induce rapid increases in alcohol self-administration. We used high-density oligonucleotide microarrays to investigate gene expression changes during chronic intermittent alcohol exposure in three brain regions that receive mesocorticolimbic dopaminergic projections and that are believed to be involved in alcohol's reinforcing actions: the medial prefrontal cortex, the nucleus accumbens and the amygdala. An independent replication of the experiment was used for RT-PCR validation of the microarray results. The protein kinase A inhibitor alpha (PKI-alpha, Pkia), a member of the endogenous PKI family implicated in reducing nuclear PKA activity, was found to be increased in all three regions tested. Conversely, we observed a downregulation of the expression of several PKA-regulated transcripts in one or more of the brain regions studied, including the activity and neurotransmitter-regulated early gene (Ania) - 1, -3, -7, -8, the transcription factors Egr1 and NGFI-B (Nr4a1) and the neuropeptide NPY. Reduced expression of PKA-regulated genes in mesocorticolimbic projection areas may have motivational significance in the rapid increase in alcohol self-administration induced by intermittent alcohol exposure.
Collapse
Affiliation(s)
- Vez Repunte-Canonigo
- Molecular and Integrative Neuroscience Department and San Diego Alcohol Research Center, The Scripps Research Institute, La Jolla, CA, 92103, USA
| | - Robert Lutjens
- Molecular and Integrative Neuroscience Department and San Diego Alcohol Research Center, The Scripps Research Institute, La Jolla, CA, 92103, USA
| | - Lena D. van der Stap
- Molecular and Integrative Neuroscience Department and San Diego Alcohol Research Center, The Scripps Research Institute, La Jolla, CA, 92103, USA
| | - Pietro Paolo Sanna
- Molecular and Integrative Neuroscience Department and San Diego Alcohol Research Center, The Scripps Research Institute, La Jolla, CA, 92103, USA
- To whom correspondence should be addressed ()
| |
Collapse
|
25
|
Kaphzan H, O'Riordan KJ, Mangan KP, Levenson JM, Rosenblum K. NMDA and dopamine converge on the NMDA-receptor to induce ERK activation and synaptic depression in mature hippocampus. PLoS One 2006; 1:e138. [PMID: 17205142 PMCID: PMC1762427 DOI: 10.1371/journal.pone.0000138] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Accepted: 12/07/2006] [Indexed: 01/19/2023] Open
Abstract
The formation of enduring internal representation of sensory information demands, in many cases, convergence in time and space of two different stimuli. The first conveys the sensory input, mediated via fast neurotransmission. The second conveys the meaning of the input, hypothesized to be mediated via slow neurotransmission. We tested the biochemical conditions and feasibility for fast (NMDA) and slow (dopamine) neurotransmission to converge on the Mitogen Activated Protein Kinase signaling pathways, crucial in several forms of synaptic plasticity, and recorded its effects upon synaptic transmission. We detected differing kinetics of ERK2 activation and synaptic strength changes in the CA1 for low and high doses of neurotransmitters in hippocampal slices. Moreover, when weak fast and slow inputs are given together, they converge on ERK2, but not on p38 or JNK, and induce strong short-term synaptic depression. Surprisingly, pharmacological analysis revealed that a probable site of such convergence is the NMDA receptor itself, suggesting it serves as a detector and integrator of fast and slow neurotransmission in the mature mammalian brain, as revealed by ERK2 activation and synaptic function.
Collapse
Affiliation(s)
- Hanoch Kaphzan
- Center for Brain and Behavior, Department of Neurobiology and Ethology, Haifa University, Haifa, Israel
| | - Kenneth J. O'Riordan
- Department of Pharmacology and the Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Kile P. Mangan
- Department of Pharmacology and the Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Jonathan M. Levenson
- Department of Pharmacology and the Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Kobi Rosenblum
- Center for Brain and Behavior, Department of Neurobiology and Ethology, Haifa University, Haifa, Israel
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
26
|
Zaitseva T, Schultz S, Schears G, Pastuszko P, Markowitz S, Greeley W, Wilson DF, Pastuszko A. Regulation of brain cell death and survival after cardiopulmonary bypass. Ann Thorac Surg 2006; 82:2247-53. [PMID: 17126142 DOI: 10.1016/j.athoracsur.2006.06.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Revised: 06/03/2006] [Accepted: 06/06/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND This study investigated the effect of low flow cardiopulmonary bypass, circulatory arrest, and selective cerebral perfusion on expression and phosphorylation of selected regulators of cell death and survival in striatum of newborn piglets. METHODS Animals were assigned to sham operation and three experimental groups. The experimental groups were placed on bypass, cooled to 18 degrees C, and subjected to 90 minutes of deep hypothermic circulatory arrest (DHCA), low-flow cardiopulmonary bypass (LFCPB) at mL/(kg x min), or selective cerebral perfusion (SCP) at 20 mL/(kg x min), followed by rewarming and 2 hours of recovery. The oxygen pressure in the microcirculation of the cortex was measured by quenching of phosphorescence. Levels of phosphorylated and total protein were determined by Western blot analysis. RESULTS Control oxygen pressure was 55 +/- 9 mm Hg and decreased during DHCA, LFCPB, and SCP to 1.1 +/- 0.6 mm Hg, 9.8 +/- 2.3 mm Hg, and 9.3 +/- 1.9 mm Hg, respectively (p < 0.001). After DHCA, N-terminal of Bcl-2-associated X protein (N-Bax) levels increased (295% +/- 15%, p < 0.01), B-cell leukemia protein (Bcl-2) levels decreased (31% +/- 9%, p < 0.01), and phosphorylation level of protein kinase B (pAkt) and extracellular signal-regulated kinase 1/2 (pERK1/2) did not change. After LFCPB and SCP, N-Bax and Bcl-2 levels were unchanged, pAkt levels increased (367% +/- 122%, p < 0.05 and 337% +/- 47%, p < 0.01, respectively), pERK1 (484% +/- 70% and 501% +/- 255%, respectively; p < 0.01) and pERK2 (569% +/- 128%; p < 0.001 and 494% +/- 162%; p < 0.05, respectively) levels increased, and total ERK2 levels also increased (279% +/- 90% and 153% +/- 44%, respectively, p < 0.05). CONCLUSIONS Stable levels of Bcl-2 and Bax and the increases in pAkt and pERK1/2 after LFCPB and SCP are likely indicators of improved chances for cell survival.
Collapse
Affiliation(s)
- Tatiana Zaitseva
- Department of Biochemistry and Biophysics, The University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Kasim S, Blake BL, Fan X, Chartoff E, Egami K, Breese GR, Hess EJ, Jinnah HA. The role of dopamine receptors in the neurobehavioral syndrome provoked by activation of L-type calcium channels in rodents. Dev Neurosci 2006; 28:505-17. [PMID: 17028428 PMCID: PMC2951315 DOI: 10.1159/000095113] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Accepted: 11/16/2005] [Indexed: 11/19/2022] Open
Abstract
In rodents, activation of L-type calcium channels with +/-BayK 8644 causes an unusual behavioral syndrome that includes dystonia and self-biting. Prior studies have linked both of these behaviors to dysfunction of dopaminergic transmission in the striatum. The current studies were designed to further elucidate the relationship between +/-BayK 8644 and dopaminergic transmission in the expression of the behavioral syndrome. The drug does not appear to release presynaptic dopamine stores, since microdialysis of the striatum revealed dopamine release was unaltered by +/-BayK 8644. In addition, the behaviors were preserved or even exaggerated in mice or rats with virtually complete dopamine depletion. On the other hand, pretreatment of mice with D(3) or D(1/5) dopamine receptor antagonists attenuated the behavioral effects of +/-BayK 8644, while pretreatment with D(2) or D(4) antagonists had no effect. In D(3) receptor knockout mice, +/-BayK 8644 elicited both dystonia and self-biting, but these behaviors were less severe than in matched controls. In D(1) receptor knockout mice, behavioral responses to +/-BayK 8644 appeared exaggerated. These results argue that the behavioral effects of +/-BayK 8644 are not mediated by a presynaptic influence. Instead, the behaviors appear to result from a postsynaptic activation of the drug, which does not require but can be modified by D(3) or D(1/5) receptors.
Collapse
MESH Headings
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology
- Animals
- Calcium Channel Agonists/pharmacology
- Calcium Channels, L-Type/drug effects
- Calcium Channels, L-Type/metabolism
- Central Nervous System Stimulants/pharmacology
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Corpus Striatum/physiopathology
- Disease Models, Animal
- Dopamine/metabolism
- Dopamine Antagonists/pharmacology
- Dystonia/chemically induced
- Dystonia/metabolism
- Dystonia/physiopathology
- Female
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Rats
- Rats, Sprague-Dawley
- Receptors, Dopamine/drug effects
- Receptors, Dopamine/genetics
- Receptors, Dopamine/metabolism
- Receptors, Dopamine D1/drug effects
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D3/drug effects
- Receptors, Dopamine D3/genetics
- Receptors, Dopamine D3/metabolism
- Self-Injurious Behavior/chemically induced
- Self-Injurious Behavior/metabolism
- Self-Injurious Behavior/physiopathology
- Synaptic Membranes/drug effects
- Synaptic Membranes/genetics
- Synaptic Membranes/metabolism
- Synaptic Transmission/drug effects
- Synaptic Transmission/physiology
- Syndrome
Collapse
Affiliation(s)
- Suhail Kasim
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Blake BL, Muehlmann AM, Egami K, Breese GR, Devine DP, Jinnah HA. Nifedipine suppresses self-injurious behaviors in animals. Dev Neurosci 2006; 29:241-50. [PMID: 17047321 PMCID: PMC2951318 DOI: 10.1159/000096414] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Accepted: 04/12/2006] [Indexed: 11/19/2022] Open
Abstract
Self-injurious behavior is a common problem in many developmental disorders. The neurobiology of this behavior is not well understood, but the differing behavioral manifestations and associations with different disorders suggest that the underlying biological mechanisms are heterogeneous. The behavioral and biological heterogeneity is also evident in several animal models, where different manifestations can be provoked under different experimental conditions. Identifying commonalities among the different mechanisms is likely to be helpful in the design of treatments useful for the broadest populations of patients. The current studies reveal that nifedipine suppresses self-injurious behavior in 4 unrelated animal models: acute administration of high doses of +/-BayK 8644 or methamphetamine in mice, dopamine agonist treatment in rats with lesions of dopamine pathways during early development and repeated administration of pemoline in rats. The effect of nifedipine does not appear to be due to nonspecific mechanisms, such as sedation, since other classes of behaviors are unaffected or exaggerated. These results suggest that nifedipine may target a common biological mechanism in the expression of self-injurious behavior, and they suggest it should be considered in the treatment of self-injury in humans.
Collapse
MESH Headings
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester
- Animals
- Behavior, Animal/drug effects
- Calcium Channel Agonists
- Calcium Channel Blockers/pharmacology
- Central Nervous System Stimulants
- Disease Models, Animal
- Female
- Male
- Methamphetamine
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Nifedipine/pharmacology
- Oxidopamine
- Pemoline
- Pregnancy
- Rats
- Rats, Long-Evans
- Rats, Sprague-Dawley
- Self-Injurious Behavior/chemically induced
- Self-Injurious Behavior/drug therapy
- Sympatholytics
Collapse
Affiliation(s)
- Bonita L Blake
- Department of Psychiatry, University of North Carolina, Chapel Hill, USA
| | | | | | | | | | | |
Collapse
|
29
|
Brown P, Gerfen CR. Plasticity within striatal direct pathway neurons after neonatal dopamine depletion is mediated through a novel functional coupling of serotonin 5-HT2 receptors to the ERK 1/2 map kinase pathway. J Comp Neurol 2006; 498:415-30. [PMID: 16871540 PMCID: PMC2585776 DOI: 10.1002/cne.21034] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dysfunction within the striatal direct and indirect projecting systems arises after 6-hydroxydopamine (6-OHDA)-induced dopamine depletion, highlighting the central regulatory function of dopamine in motor systems. However, the striatal 5-hydroxytryptamine (5-HT) innervation remains intact after 6-OHDA lesions, suggesting that the 5-HT system may contribute to the lesion-induced dysfunction, or alternatively, it may adapt and compensate for the dopamine deficit. Neonatal 6-OHDA lesions actually give rise to a 5-HT axonal hyperinnervation within the dorsal striatum, further reinforcing the idea that the 5-HT system plays a central role in striatal function after dopamine depletion. Here we show that neonatal but not adult 6-OHDA lesions result in a novel coupling of 5-HT2 receptors to the ERK1/2/MAP Kinase pathway, a signaling cascade known to regulate neuronal plasticity. Chloroamphetamine-induced 5-HT release or direct stimulation of striatal 5-HT2 receptors via the 5-HT2 agonist DOI, produced robust ERK1/2 phosphorylation throughout the dorsal striatum of neonatal lesioned animals, a response not observed within the intact striatum. Pretreatment with the select 5-HT2 receptor antagonist Ketanserin blocked DOI-induced ERK1/2 phosphorylation. This drug-induced ERK1/2 phosphorylation was subsequently shown to be restricted to direct pathway striatal neurons. Our data show that adaptation of direct pathway neurons after neonatal 6-OHDA lesions involves coupling of 5-HT2 receptors to the ERK1/2/MAP Kinase cascade, a pathway not typically active in these neurons. Because dopamine-mediated signaling is redundant after 6-OHDA lesions, 5-HT-mediated stimulation of the ERK1/2/MAP Kinase pathway may provide an alternative signaling route allowing the regulation of neuronal gene expression and neuronal plasticity in the absence of dopamine.
Collapse
Affiliation(s)
- Pierre Brown
- Laboratory of Systems Neuroscience, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
30
|
Abstract
Glutamate receptors regulate gene expression in neurons by activating intracellular signaling cascades that phosphorylate transcription factors within the nucleus. The mitogen-activated protein kinase (MAPK) cascade is one of the best characterized cascades in this regulatory process. The Ca(2+)-permeable ionotropic glutamate receptor, mainly the NMDA receptor subtype, activates MAPKs through a biochemical route involving the Ca(2+)-sensitive Ras-guanine nucleotide releasing factor, Ca(2+)/calmodulin-dependent protein kinase II, and phosphoinositide 3-kinase. The metabotropic glutamate receptor (mGluR), however, activates MAPKs primarily through a Ca(2+)-insensitve pathway involving the transactivation of receptor tyrosine kinases. The adaptor protein Homer also plays a role in this process. As an information superhighway between surface glutamate receptors and transcription factors in the nucleus, active MAPKs phosphorylate specific transcription factors (Elk-1 and CREB), and thereby regulate distinct programs of gene expression. The regulated gene expression contributes to the development of multiple forms of synaptic plasticity related to long-lasting changes in memory function and addictive properties of drugs of abuse. This review, by focusing on new data from recent years, discusses the signaling mechanisms by which different types of glutamate receptors activate MAPKs, features of each MAPK cascade in regulating gene expression, and the importance of glutamate/MAPK-dependent synaptic plasticity in memory and addiction.
Collapse
Affiliation(s)
- John Q Wang
- Department of Basic Medical Science, University of Missouri-Kansas City, School of Medicine, Kansas City, Missouri, USA.
| | | | | |
Collapse
|
31
|
Cao JL, Liu HL, Wang JK, Zeng YM. Cross talk between nitric oxide and ERK1/2 signaling pathway in the spinal cord mediates naloxone-precipitated withdrawal in morphine-dependent rats. Neuropharmacology 2006; 51:315-26. [PMID: 16712881 DOI: 10.1016/j.neuropharm.2006.03.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 03/19/2006] [Accepted: 03/20/2006] [Indexed: 10/24/2022]
Abstract
Our recent study has shown activation of spinal extracellular signal-regulated kinase-1 and -2 (ERK1/2), a member of the mitogen-activated protein kinase (MAPK) family, contributes to naloxone-precipitated withdrawal and withdrawal-induced spinal neuronal sensitization in morphine-dependent rats. However, the mechanism and significance of the spinal ERK1/2 activation during morphine dependence and withdrawal remain unknown. In this study, we reported that intrathecal (i.t.) pretreatment with either the non-selective nitric oxide synthase (NOS) inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME), neuronal NOS (nNOS) inhibitor 7-nitro indazole (7-NI), or the inducible NOS (iNOS) inhibitor aminoguanidine (AG), could reduce morphine withdrawal-induced increase of phospho-ERK1/2 (pERK1/2) expression in the rat spinal cord. On the other hand, attenuation of the spinal ERK phosphorylation by the MAPK kinase (MEK) inhibitor U0126 also could inhibit the increase of nNOS and iNOS expression in the spinal cord of morphine withdrawal rats. Inhibitory expression of pERK1/2 by i.t. NOS inhibitor L-NAME, 7-NI or AG and of nNOS and iNOS by i.t. U0126 in the spinal cord were accompanied by decreased scores of morphine withdrawal and the inhibited spinal Fos protein (a maker for neuronal excitation or activation) expression induced by morphine withdrawal. These findings suggest cross talk between nitric oxide (NO) and the ERK1/2 signaling pathway mediates morphine withdrawal and withdrawal-induced spinal neuronal sensitization in morphine-dependent rats.
Collapse
Affiliation(s)
- Jun-Li Cao
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical College, Jiangsu Institute of Anesthesiology, Jiangsu Province, Xuzhou, Jiangsu 221002, PR China.
| | | | | | | |
Collapse
|
32
|
Beard JL, Felt B, Schallert T, Burhans M, Connor JR, Georgieff MK. Moderate iron deficiency in infancy: Biology and behavior in young rats. Behav Brain Res 2006; 170:224-32. [PMID: 16569441 DOI: 10.1016/j.bbr.2006.02.024] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Revised: 01/13/2006] [Accepted: 02/20/2006] [Indexed: 11/17/2022]
Abstract
Iron deficiency anemia in early childhood is associated with developmental delays and perhaps, irreversible alterations in neurological functioning. The goals were to determine if dietary induced gestational and lactational iron deficiency alters brain monoamine metabolism and behaviors dependent on that neurotransmitter system. Young pregnant rats were provided iron deficient or control diets from early in gestation through to weaning of pups and brain iron concentration, regional monoamine variables and achievement of specific developmental milestones were determined throughout lactation. Despite anemia during lactation, most brain iron concentrations did not fall significantly until P25, and well after significant changes in monoamine levels, transporter levels, and D2R density changed in terminal fields. The changes in D2R density were far smaller than previously observed models that utilized severe dietary restriction during lactation or after weaning. Iron deficient pups had normal birth weight, but were delayed in the attainment of a number of milestones (bar holding, vibrissae-evoked forelimb placing). This approach of iron deficiency in utero and during lactation sufficient to cause moderate anemia but not stunt growth demonstrates that monaminergic metabolism changes occur prior to profound declines in brain iron concentration and is associated with developmental delays. Similar developmental delays in iron deficient human infants suggest to us that alterations in iron status during this developmental period likely affects developing brain monaminergic systems in these infants.
Collapse
MESH Headings
- Age Factors
- Anemia, Iron-Deficiency/etiology
- Anemia, Iron-Deficiency/metabolism
- Anemia, Iron-Deficiency/physiopathology
- Animals
- Animals, Newborn/physiology
- Animals, Newborn/psychology
- Behavior, Animal/physiology
- Biogenic Monoamines/metabolism
- Brain/metabolism
- Brain/pathology
- Disease Models, Animal
- Female
- Gene Expression Regulation, Developmental/physiology
- Hematocrit/methods
- Iron/blood
- Iron Deficiencies
- Lactation/physiology
- Male
- Pregnancy
- Rats
- Rats, Sprague-Dawley
- Receptors, Dopamine D2/metabolism
Collapse
Affiliation(s)
- John L Beard
- Department of Nutrition, Penn State University, PA 16802, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Cao JL, He JH, Ding HL, Zeng YM. Activation of the spinal ERK signaling pathway contributes naloxone-precipitated withdrawal in morphine-dependent rats. Pain 2005; 118:336-349. [PMID: 16289800 DOI: 10.1016/j.pain.2005.09.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Revised: 08/14/2005] [Accepted: 09/02/2005] [Indexed: 11/22/2022]
Abstract
Extracellular signal-regulated kinase (ERK), a mitogen-activated protein kinases (MAPK), transduces a broad range of extracellular stimuli into diverse intracellular responses. Recent studies have showed that ERK activation in the supraspinal level involved in the development of drug dependence, especially in psychological dependence. In this study, we reported that the spinal ERK signaling pathway was activated by chronic morphine injection. There was a further increase in ERK activation after naloxone-precipitated withdrawal. Furthermore, attenuation of the spinal ERK phosphorylation by intrathecal a MAPK kinase (MEK) inhibitor U0126 or knockdown of the spinal ERK by antisense oligonucleotides not only decreased the scores of morphine withdrawal, but also attenuated withdrawal-induced allodynia, which were accompanied by decreased ERK phosphorylation in the spinal cord. The spinal ERK inhibition or knockdown also reduced morphine withdrawal-induced phosphorylation of cAMP response element binding protein (CREB), which is one of the important downstream substrates of ERK pathway, and Fos expression. The involvement of the spinal ERK in morphine withdrawal was supported by our finding that intrathecal N-methyl-D-aspartate receptor antagonist MK-801 or protein kinase C inhibitor chelerythrine chloride suppressed withdrawal-induced ERK activation in the spinal cord and attenuated morphine withdrawal symptoms. These findings suggest activation of the spinal ERK signaling pathway contributes naloxone-precipitated withdrawal in morphine-dependent rats.
Collapse
Affiliation(s)
- Jun-Li Cao
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical College, 99 Huaihai West Road, Xuzhou 221002, People's Republic of China Jiangsu Key Laboratory of Anesthesiology, Jiangsu Institute of Anesthesiology, 99 Huaihai West Road, Xuzhou 221002, People's Republic of China Department of Physiology, University of Texas, Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX 75390-9040, USA
| | | | | | | |
Collapse
|
34
|
Campbell BM, Kreipke CW, Walker PD. Failure of MK-801 to suppress D1 receptor-mediated induction of locomotor activity and striatal preprotachykinin mRNA expression in the dopamine-depleted rat. Neuroscience 2005; 137:505-17. [PMID: 16289829 DOI: 10.1016/j.neuroscience.2005.09.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Revised: 08/26/2005] [Accepted: 09/21/2005] [Indexed: 11/26/2022]
Abstract
N-methyl-D-aspartate receptor antagonism exerts suppressive influences over dopamine D1 receptor-mediated striatal gene expression and locomotor behavior in the intact rat. The present study examined the effects of the N-methyl-D-aspartate receptor antagonist MK-801 on locomotor activity and striatal preprotachykinin mRNA expression stimulated by the D1 agonist (+/-)6-chloro-7, 8-dihydroxy-3-allyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrobromide in rats with bilateral dopamine lesions. Two months after neonatal dopamine lesions with 6-hydroxydopamine, rats were challenged with (+/-)6-chloro-7, 8-dihydroxy-3-allyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrobromide (1.0 mg/kg) 15 min after administration of the N-methyl-D-aspartate receptor antagonist MK-801 (0.1 mg/kg). In the intact rat, MK-801 prevented the induction of striatal preprotachykinin mRNA by D1 agonism. Similarly, direct infusion of (+/-)6-chloro-7, 8-dihydroxy-3-allyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrobromide (3.0 microg) into the intact striatum produced an increase in locomotor activity that was suppressed by MK-801 (1.0 microg) co-infusion. In the dopamine-depleted rat, MK-801 (0.1 mg/kg) administered prior to (+/-)6-chloro-7, 8-dihydroxy-3-allyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrobromide (1.0 mg/kg) increased, rather than suppressed, striatal preprotachykinin mRNA levels. Intrastriatal infusion of MK-801 (1.0 microg) failed to inhibit D1-mediated induction of motor activity in dopamine-depleted animals. Together, these data provide further support that N-methyl-D-aspartate receptor antagonists lose their ability to block D1-mediated behavioral activation following dopamine depletion. The activation, rather than suppression, of tachykinin neurons of the direct striatonigral pathway may play a facilitatory role in this mechanism.
Collapse
Affiliation(s)
- B M Campbell
- Cellular and Clinical Neurobiology Program, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | |
Collapse
|
35
|
Jenab S, Festa ED, Nazarian A, Wu HBK, Sun WL, Hazim R, Russo SJ, Quinones-Jenab V. Cocaine induction of ERK proteins in dorsal striatum of Fischer rats. ACTA ACUST UNITED AC 2005; 142:134-8. [PMID: 16271798 DOI: 10.1016/j.molbrainres.2005.08.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2005] [Revised: 07/27/2005] [Accepted: 08/17/2005] [Indexed: 11/24/2022]
Abstract
Cocaine is an addictive psychostimulant that induces fos and opioid gene expression by activating the dopamine receptors and the PKA pathways in dopamine D1 and a glutamate NMDA-dependent mechanisms in the striatum. In this study, we show that a single cocaine administration induces ERK phosphorylation in the caudate/putamen of Fischer rats. This increase in Phospho-ERK is diminished by pre-administration of SCH23390, or MK801 but not with pre-administration of eticlopride. Furthermore, this single cocaine administration does not alter the levels of phospho-CREB protein or CREB-DNA bindings in the caudate/putamen protein extracts but does increase phospho-Elk-1 protein levels in the same extracts.
Collapse
Affiliation(s)
- Shirzad Jenab
- Department of Psychology, Hunter College of The City University of New York, NY 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Choe ES, Shin EH, Wang JQ. Inhibition of protein phosphatase 2B upregulates serine phosphorylation of N-methyl-d-aspartate receptor NR1 subunits in striatal neurons in vivo. Neurosci Lett 2005; 384:38-43. [PMID: 15890444 DOI: 10.1016/j.neulet.2005.04.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2005] [Revised: 04/14/2005] [Accepted: 04/15/2005] [Indexed: 11/27/2022]
Abstract
This study investigated the role of protein phosphatase 2B (calcineurin) in regulating phosphorylation of N-methyl-D-aspartate receptor (NMDAR) NR1 subunits and other phosphoproteins in the rat striatum in vivo. In chronically cannulated rats, microinjection of the calcineurin selective inhibitor cyclosporin A increased phosphorylation of NMDAR NR1 subunits at serine 896 and serine 897 in the injected dorsal striatum. The increase in NMDAR NR1 phosphorylation was dose-dependent in a dose range surveyed (0.005, 0.05, and 0.5 nmol). Parallel with increased serine phosphorylation of NR1 subunits, cyclosporin A dose-dependently increased phosphorylation of a Ca2+-sensitive protein kinase, extracellular signal-regulated protein kinase 1/2 (ERK1/2), and a Ca2+/cAMP-sensitive transcription factor, cAMP response element-binding protein (CREB), in the dorsal striatum. Using an immediate early gene product Fos as a reporter of inducible gene expression, cyclosporin A was found to upregulate Fos expression in the dorsal striatum. These results indicate that calcineurin plays an important role in the tonic dephosphorylation of NMDAR NR1 subunits and other two key cytoplasmic and nuclear signaling proteins (ERK1/2 and CREB) in striatal neurons.
Collapse
Affiliation(s)
- Eun Sang Choe
- Department of Biology, Pusan National University, 30 Jangjeon-dong, Kumjeong-gu, Pusan 609-735, Republic of Korea.
| | | | | |
Collapse
|
37
|
Breese GR, Knapp DJ, Criswell HE, Moy SS, Papadeas ST, Blake BL. The neonate-6-hydroxydopamine-lesioned rat: a model for clinical neuroscience and neurobiological principles. ACTA ACUST UNITED AC 2005; 48:57-73. [PMID: 15708628 DOI: 10.1016/j.brainresrev.2004.08.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2004] [Indexed: 01/08/2023]
Abstract
In 1973, a technique of administering 6-hydroxydopamine (2,4,5-trihydroxyphenylethylamine) intracisternally to neonate rats was introduced to selectively reduce brain dopamine (neonate-lesioned rat). This neonate treatment proved unique when compared to rats lesioned as adults with 6-hydroxydopamine--prompting the discovery of differing functional characteristics resulting from the age at which brain dopamine is reduced. A realization was that neonate-lesioned rats modeled the loss of central dopamine and the increased susceptibility for self-injury in Lesch-Nyhan disease, which allowed identification of drugs useful in treating self-injury in mentally retarded patients. The neonate-lesioned rat has also been proposed to model the hyperactivity observed in attention-deficit hyperactivity disorder. Because the neonate-lesioned rat exhibits enhanced sensitization to repeated NMDA receptor antagonist administration and has functional changes characteristic of schizophrenia, the neonate lesioning is believed to emulate the hypothesized NMDA hypofunction in this psychiatric disorder. Besides modeling features of neurological and psychiatric disorders, important neurobiological concepts emerged from pharmacological studies in the neonate-lesioned rats. One was the discovery of coupling of D1/D2-dopamine receptor function. Another was the progressive increase in responsiveness to repeated D1-dopamine agonist administration referred to as "priming" of D1-dopamine receptor function. Additionally, a unique profile of signaling protein expression related to neonate reduction of dopamine has been identified. Thus, from modeling characteristics of disease to defining adaptive mechanisms related to neonatal loss of dopamine, the neonate-lesioned rat has had a persisting influence on neuroscience. Despite an extraordinary legacy from studies of the neurobiology of this treatment, a host of unknowns remain that will inspire future investigations.
Collapse
Affiliation(s)
- George R Breese
- Department of Psychiatry, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7178, USA.
| | | | | | | | | | | |
Collapse
|