251
|
Jin DQ, Lim CS, Sung JY, Choi HG, Ha I, Han JS. Ulva conglobata, a marine algae, has neuroprotective and anti-inflammatory effects in murine hippocampal and microglial cells. Neurosci Lett 2006; 402:154-8. [PMID: 16644126 DOI: 10.1016/j.neulet.2006.03.068] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2006] [Revised: 03/27/2006] [Accepted: 03/29/2006] [Indexed: 11/22/2022]
Abstract
It has been reported that inflammatory processes are associated with the pathophysiology of Alzheimer's disease (AD), and the treatment of AD using anti-inflammatory agents slows the progress of AD. Marine algae have been utilized in food products as well as in medicine products for a variety of purposes. In this study, we investigated the neuroprotective effects of methanol extracts of Ulva conglobata (U. conglobata), a marine algae, on glutamate-induced neurotoxicity in the murine hippocampal HT22 cell line and the anti-inflammatory effects on interferon gamma (IFN-gamma)-induced microglial activation in BV2 cells. U. conglobata methanol extracts significantly attenuated the neurotoxicity induced by glutamate in HT22 cells and inhibited nitric oxide production induced by IFN-gamma in BV2 cells. U. conglobata methanol extract treatments were also examined and it was found that they almost completely suppressed the expression of the proinflammatory enzyme cyclooxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS). These results suggest that U. conglobata possesses therapeutic potential for combating neurodegenerative diseases associated with neuroinflammation.
Collapse
Affiliation(s)
- Da-Qing Jin
- Graduate Program in Neuroscience, Institute for Brain Science and Technology (IBST), Hanwha CC R&D Center Building, Inje University, 6 Shinsung-dong, Yuseong-gu, Daejeon 305-804, South Korea
| | | | | | | | | | | |
Collapse
|
252
|
Weerasinghe GR, Coon SL, Bhattacharjee AK, Harry GJ, Bosetti F. Regional protein levels of cytosolic phospholipase A2 and cyclooxygenase-2 in Rhesus monkey brain as a function of age. Brain Res Bull 2006; 69:614-21. [PMID: 16716827 PMCID: PMC1473171 DOI: 10.1016/j.brainresbull.2006.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Revised: 12/30/2005] [Accepted: 03/06/2006] [Indexed: 10/24/2022]
Abstract
Limited evidence suggests that brain cytosolic phospholipase A(2) (cPLA(2)), which selectively releases arachidonic acid (AA) from membrane phospholipids, and cyclooxygenase-2 (COX-2), the rate-limiting enzyme for AA metabolism to prostanoids, change as a function of normal aging. In this study, we examined the protein levels of cPLA(2) and COX-2 enzymes in hippocampus, frontal pole and cerebellum from young (2-5 years old), middle-aged (8-11 years old) and old (23 years old) male and female Rhesus monkeys. In the cerebellum, cPLA(2) protein level was higher in the young brain as compared to levels seen at both middle-aged and old. Similarly, in the frontal pole, the young brain showed a higher level of COX-2 protein as compared to the levels seen at both older ages. For both, once an animal reached 8-11 years of age the levels appeared to remain relatively constant over the next decade. Immunohistochemistry of COX-2 protein within the brain demonstrated no significant change in the localization to neurons within the frontal pole. Qualitatively, a greater number of neurons were positively stained for COX-2 in the young brain than in the aged brain. Based on the previous reports of localization of cPLA(2) and COX-2 at post-synaptic sites in neurons results from the current study suggest that the elevated protein levels of the two enzymes seen in the younger brain is related to the greater potential for synaptic plasticity across multiple neurons as a function of age and that cPLA(2) and COX-2 may be considered as post-synaptic markers.
Collapse
Affiliation(s)
- Gayani R. Weerasinghe
- Brain Physiology and Metabolism Section, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Steven L. Coon
- Section on Neuroendocrinology, National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | | | - G. Jean Harry
- Neurotoxicology Group, National Institute of Environmental Health Sciences , NIH, Research Triangle Park, NC 27709, USA
| | - Francesca Bosetti
- Brain Physiology and Metabolism Section, National Institute on Aging, NIH, Bethesda, MD 20892, USA
- *Corresponding author: Brain Physiology and Metabolism Section, NIA, NIH; 9 Memorial Drive, Bldg. 9, Rm. 1S126, Bethesda, MD 20892-0947, USA. Tel: (301) 594-5077; Fax: (301) 402-0074; E-mail:
| |
Collapse
|
253
|
Kim DK, Jang TJ. Cyclooxygenase-2 expression and effect of celecoxib in flurothyl-induced neonatal seizure. Int J Exp Pathol 2006; 87:73-8. [PMID: 16436115 PMCID: PMC2517343 DOI: 10.1111/j.0959-9673.2006.00457.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Endogenous PGE(2) dynamically regulates membrane excitability, synaptic transmission and plasticity. Neonatal seizures are associated with a number of activity-dependent changes in brain development including altered synaptogenesis and synaptic plasticity as well as reduction in neurogenesis. Thus, it is reasonable to hypothesize that alteration of cyclooxygenase-2 (COX-2) expression induced by neonatal seizure may influence brain development. We evaluated the expression of COX-2 and microsomal prostaglandin E synthase (mPGES) by Western blot analysis and immnohistochemistry in flurothyl-induced neonatal seizure and also studied the effect of celecoxib on seizure induction. Seven to 10 days old Sprague-Dawley rats were used for control (n = 18) and experimental group (n = 30). Recurrent seizure group showed more increased level of COX-2 expression than control group. However, the level of mPGES-2 expression was similar in both groups, and mPGES-1 was not detected. Hippocampus of control rats showed endogenous COX-2 expression, which was localized mainly in CA3 region. This localization pattern was similar in recurrent seizure rats, but intensity of COX-2 expression was more increased than in control rats. Celecoxib treatment significantly delayed the seizure attack and also reduced COX-2 expression. In conclusion, this study suggests that COX-2 expression is related to epileptogenesis in flurothyl-induced neonatal seizure model and shows the possibility that its inhibition lessens functional impairments that occurred in neonatal seizure.
Collapse
Affiliation(s)
- Doo Kwun Kim
- Department of Pediatrics, Dongguk University College of MedicineKyongju, Kyongbuk, Korea
| | - Tae Jung Jang
- Department of Pathology, Dongguk University College of MedicineKyongju, Kyongbuk, Korea
| |
Collapse
|
254
|
Sharifzadeh M, Tavasoli M, Soodi M, Mohammadi-Eraghi S, Ghahremani MH, Roghani A. A time course analysis of cyclooxygenase-2 suggests a role in spatial memory retrieval in rats. Neurosci Res 2006; 54:171-9. [PMID: 16364481 DOI: 10.1016/j.neures.2005.11.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Revised: 10/22/2005] [Accepted: 11/10/2005] [Indexed: 11/21/2022]
Abstract
We previously showed a role for COX-2 in spatial memory retention. In that study we investigated the effects of post-training intrahippocampal infusion of celecoxib as a COX-2-specific inhibitor on spatial memory retention. Those infusions impaired spatial memory retention in the Morris water maze. In the present study a time course analysis of role of COX-2 in spatial memory was conducted. Here stereotaxic surgery was employed for the bilateral implantation of guide cannulas into the CA1 region of the hippocampus. Training trials were started after recovery of the animals. Immediately after last trial of training on third day, the celecoxib (0.1M) was infused bilaterally and testing trials, were performed 1, 2, 3, and 7 days after celecoxib infusions. Significant alterations were observed in escape latency and traveled distance 2 and 3 days after celecoxib infusions. The maximum impairment was obtained 72 h after the infusions. The data suggests that the effect of celecoxib is transient and that its effect on performance is likely caused by a problem in memory retrieval. Quantification analyses of the immunostaining of COX-2-containing neurons in the dorsal hippocampus show that celecoxib infusions significantly reduced (P<0.05) COX-2 immunoreactivity for the animals that were tested 3 days after the drug infusion. Results from the behavioral study along with the findings from immunohistochemical analyses suggest that COX-2 has significant role in spatial memory retrieval. Moreover, the memory deficits induced by the infusions continuously persists for 3 days.
Collapse
Affiliation(s)
- Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Pharmaceutical Sciences and Medicinal Plants Research Centers, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | |
Collapse
|
255
|
HaMai D, Rinderknecht AL, Guo-Sharman K, Kleinman MT, Bondy SC. Decreased expression of inflammation-related genes following inhalation exposure to manganese. Neurotoxicology 2006; 27:395-401. [PMID: 16476481 DOI: 10.1016/j.neuro.2005.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Revised: 10/29/2005] [Accepted: 11/09/2005] [Indexed: 11/17/2022]
Abstract
Excessive exposure to manganese (Mn) by inhalation can induce psychosis and Parkinsonism. The clinical manifestations of Mn neurotoxicity have been related to numerous physiological and cellular processes, most notably dopamine depletion. However, few studies have explored the molecular events that are triggered in response to exposure to Mn by inhalation. In this current study, the transcriptional patterns of genes related to oxidative stress or inflammation were examined in the brain rats of exposed to inhaled Mn during either gestation or early adulthood. The expression of genes encoding for proteins critical to an inflammatory response and/or possessing pro-oxidant properties, including TGFbeta and nNOS, were slightly depressed by prenatal exposure, whereas inhalation exposure to Mn during adulthood markedly down-regulated their transcription. However, when exposures to manganese occurred during gestation, the extent of altered gene expression induced by subsequent exposure to Mn in adulthood was reduced. This suggests that prior exposure to Mn may have attenuated the effects of inhalation exposure to Mn in adulthood, in which the expression of inflammation-related genes were suppressed.
Collapse
Affiliation(s)
- Diem HaMai
- Department of Community & Environmental Medicine, University of California, Irvine, CA 92697-1825, USA.
| | | | | | | | | |
Collapse
|
256
|
Candelario-Jalil E, Slawik H, Ridelis I, Waschbisch A, Akundi RS, Hüll M, Fiebich BL. Regional distribution of the prostaglandin E2 receptor EP1 in the rat brain: accumulation in Purkinje cells of the cerebellum. J Mol Neurosci 2006; 27:303-10. [PMID: 16280600 DOI: 10.1385/jmn:27:3:303] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2005] [Accepted: 06/14/2005] [Indexed: 11/11/2022]
Abstract
Prostaglandin E2 (PGE2), is a major prostanoid produced by the activity of cyclooxygenases (COX) in response to various physiological and pathological stimuli. PGE2 exerts its effects by activating four specific E-type prostanoid receptors (EP1, EP2, EP3, and EP4). In the present study, we analyzed the expression of the PGE2 receptor EP1 (mRNA and protein) in different regions of the adult rat brain (hippocampus, hypothalamus, striatum, prefrontal cerebral cortex, parietal cortex, brain stem, and cerebellum) using reverse transcription- polymerase chain reaction, Western blotting, and immunohistochemical methods. On a regional basis, levels of EP1 mRNA were the highest in parietal cortex and cerebellum. At the protein level, we found very strong expression of EP1 in cerebellum, as revealed by Western blotting experiments. Furthermore, the present study provides for the first time evidence that the EP1 receptor is highly expressed in the cerebellum, where the Purkinje cells displayed very high immunolabeling of their perikaryon and dendrites, as observed in the immunohistochemical analysis. Results from the present study indicate that the EP1 prostanoid receptor is expressed in specific neuronal populations, which possibly determine the region-specific response to PGE2.
Collapse
|
257
|
Basselin M, Villacreses NE, Langenbach R, Ma K, Bell JM, Rapoport SI. Resting and arecoline-stimulated brain metabolism and signaling involving arachidonic acid are altered in the cyclooxygenase-2 knockout mouse. J Neurochem 2006; 96:669-79. [PMID: 16405503 DOI: 10.1111/j.1471-4159.2005.03612.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abstract Studies were performed to determine if cyclooxygenase (COX)-2 regulates muscarinic receptor-initiated signaling involving brain phospholipase A2 (PLA2) activation and arachidonic acid (AA; 20 : 4n-6) release. AA incorporation coefficients, k* (brain [1-14C]AA radioactivity/integrated plasma radioactivity), representing this signaling, were measured following the intravenous injection of [1-14C]AA using quantitative autoradiography, in each of 81 brain regions in unanesthetized COX-2 knockout (COX-2(-/-)) and wild-type (COX-2(+/+)) mice. Mice were administered arecoline (30 mg/kg i.p.), a non-specific muscarinic receptor agonist, or saline i.p. (baseline control). At baseline, COX-2(-/-) compared with COX-2(+/+) mice had widespread and significant elevations of k*. Arecoline increased k* significantly in COX-2(+/+) mice compared with saline controls in 72 of 81 brain regions, but had no significant effect on k* in any region in COX-2(-/-) mice. These findings, when related to net incorporation rates of AA from brain into plasma, demonstrate enhanced baseline brain metabolic loss of AA in COX-2(-/-) compared with COX-2(+/+) mice, and an absence of a normal k* response to muscarinic receptor activation. This response likely reflects selective COX-2-mediated conversion of PLA2-released AA to prostanoids.
Collapse
Affiliation(s)
- Mireille Basselin
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda,MD 20892-0947, USA.
| | | | | | | | | | | |
Collapse
|
258
|
Abstract
Glutamate release activates signaling pathways important for learning and memory, and over-stimulation of these pathways during seizures leads to aberrant synaptic plasticity associated with hyper-excitable, seizure-prone states. Seizures induce rapid accumulation of membrane lipid-derived fatty acids at the synapses which, evidence suggests, regulate maladaptive connectivity. Here we give an overview of the significance of the arachidonyl- and inositol-derived messengers, prostaglandins (PGs) and diacylglycerol (DAG), in experimental models of epilepsy. We use studies conducted in our own laboratory to highlight the pro-epileptogenic role of cyclooxygenase-2 (COX-2) and its products, the PGs, and we discuss the possible mechanisms by which PGs may regulate membrane excitability and synaptic transmission at the cellular level. We conclude with a discussion of AA-DAG signaling in synaptic plasticity and seizure susceptibility with an emphasis on recent studies in our laboratory involving DAG kinase epsilon (DGKepsilon)-knockout mice.
Collapse
Affiliation(s)
- Kasie K Cole-Edwards
- LSU Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA 70112, USA
| | | |
Collapse
|
259
|
|
260
|
Sang N, Zhang J, Marcheselli V, Bazan NG, Chen C. Postsynaptically synthesized prostaglandin E2 (PGE2) modulates hippocampal synaptic transmission via a presynaptic PGE2 EP2 receptor. J Neurosci 2006; 25:9858-70. [PMID: 16251433 PMCID: PMC6725559 DOI: 10.1523/jneurosci.2392-05.2005] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Increasing evidence suggests that cyclooxygenase-2 (COX-2) is involved in synaptic transmission and plasticity, and prostaglandin E2 (PGE2) is a key molecule in COX-2-meduated synaptic modification. However, the precise mechanisms, in particular, which subtypes of PGE2 receptors (EPs) mediate the PGE2-induced synaptic response, are not clear. Recently, we demonstrated that EPs are expressed heterogeneously in the hippocampus, and EP2/4 are mainly expressed in presynaptic terminals. Here, we report that PGE2 increased synaptic stimulus-evoked amplitudes of EPSPs in hippocampal slices and frequency of miniature EPSCs (mEPSCs) in hippocampal neurons in culture. These actions were mimicked by an EP2 agonist and attenuated by protein kinase A inhibitors. Decrease of EP2 expression through silencing the EP2 gene eliminated PGE2-induced increase of the frequency of mEPSCs. COX-2 and microsomal PGE synthase-1 (mPGES-1) and mPGES-2 are present in postsynaptic dendritic spines, because they are colocalized with PSD-95 (postsynaptic density-95), a postsynaptic marker. In addition, the frequency of mEPSCs was enhanced in neurons pretreated with interleukin-1beta or lipopolysaccharide, which elevated expression of COX-2 and mPGES-1 and produced PGE2, and this enhancement was inhibited by a COX-2 inhibitor that inhibited production of PGE2. Our results suggest that PGE2 synthesized by postsynaptically localized COX-2 functions as a retrograde messenger in hippocampal synaptic signaling via a presynaptic EP2 receptor.
Collapse
Affiliation(s)
- Nan Sang
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | |
Collapse
|
261
|
Abstract
The neuropathology of Alzheimer's disease (AD) is characterized by deposits of amyloid beta (Abeta) peptides and neurofibrillary tangles, but also, among other aspects, by signs of a chronic inflammatory process. Epidemiological studies have shown that long-term use of nonsteroidal antiinflammatory drugs (NSAIDs) reduces the risk of developing AD and delays its onset. The classic target of NSAIDs is the prevention of cyclooxygenase (COX) activation. The main mechanism of action of COXs is the synthesis of prostaglandins, some of which have potent inflammatory activity. The discovery of two isoforms of this enzyme, COX-1 and COX-2, and that the latter is inducible by inflammatory cytokines supported the hypothesis that its inhibition would result in a potent antiinflammatory effect and led to the rapid development of selective COX-2 inhibitors, collectively called coxibs. Based on this rationale, some coxibs have been used in clinical trials for AD patients, but all the results obtained so far have been negative. Here, we review our knowledge in terms of COX-2 in the central nervous system, COX-2 and Abeta formation, and finally COX-2 and AD pathogenesis to understand the reasons why these drugs have failed and whether there is any scientific support to keep them as therapeutic tools for this chronic disease.
Collapse
Affiliation(s)
- Omidreza Firuzi
- Department of Pharmacology, University of Pennsylvania, School of Medicine. Philadelphia, PA 19014, USA
| | | |
Collapse
|
262
|
Melnikova T, Savonenko A, Wang Q, Liang X, Hand T, Wu L, Kaufmann WE, Vehmas A, Andreasson KI. Cycloxygenase-2 activity promotes cognitive deficits but not increased amyloid burden in a model of Alzheimer’s disease in a sex-dimorphic pattern. Neuroscience 2006; 141:1149-62. [PMID: 16753269 DOI: 10.1016/j.neuroscience.2006.05.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 03/30/2006] [Accepted: 05/01/2006] [Indexed: 11/25/2022]
Abstract
Administration of non-steroidal anti-inflammatory agents reduces the risk of developing Alzheimer's disease in normal aging populations, an effect that may occur from inhibition of the cyclooxygenases, the rate-limiting enzymes in the formation of prostaglandins. In this study, we investigated whether increased activity of cyclooxygenase-2 (COX-2), the inducible isoform of cyclooxygenase, potentiates disease progression in a transgenic mouse model of Alzheimer's disease. To study the functional effects of COX-2 activity, male and female bigenic mice (amyloid precursor protein with Swedish mutation [APPswe]-presenilin-1 protein with deletion of exon 9 [PS1dE9] and trigenic COX-2/APPswe-PS1dE9) were behaviorally tested +/-administration of the selective COX-2 inhibitor celecoxib. Behavioral testing included a three-trial Y maze that measures spatial working and recognition memories and an open field task that tested levels of hyperactivity. Overexpression of COX-2 in APPswe-PS1dE9 mice resulted in specific deficits in spatial working memory in female but not male mice. These sex-specific deficits were abolished by pharmacological inhibition of COX-2 activity. Importantly, COX-2-associated deficits were dependent on co-expression of all three transgenes since COX-2 single transgenic and APPswe-PS1dE9 bigenic mice showed normal memory. Quantification of amyloid plaque load and total Abeta 40 and 42 peptides did not reveal significant differences in trigenic versus bigenic mice treated with either vehicle or celecoxib. Taken together, these data indicate an interaction between the effects of COX-2 and Abeta peptides on cognition that occurs in a sex-specific manner in the absence of significant changes in amyloid burden. These findings suggest that pathological activation of COX-2 may potentiate the toxicity of Abeta peptides, particularly in females, without significantly affecting Abeta accumulation.
Collapse
Affiliation(s)
- T Melnikova
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
263
|
Rickhag M, Wieloch T, Gidö G, Elmér E, Krogh M, Murray J, Lohr S, Bitter H, Chin DJ, von Schack D, Shamloo M, Nikolich K. Comprehensive regional and temporal gene expression profiling of the rat brain during the first 24 h after experimental stroke identifies dynamic ischemia-induced gene expression patterns, and reveals a biphasic activation of genes in surviving tissue. J Neurochem 2006; 96:14-29. [PMID: 16300643 DOI: 10.1111/j.1471-4159.2005.03508.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In order to identify biological processes relevant for cell death and survival in the brain following stroke, the postischemic brain transcriptome was studied by a large-scale cDNA array analysis of three peri-infarct brain regions at eight time points during the first 24 h of reperfusion following middle cerebral artery occlusion in the rat. K-means cluster analysis revealed two distinct biphasic gene expression patterns that contained 44 genes (including 18 immediate early genes), involved in cell signaling and plasticity (i.e. MAP2K7, Sprouty2, Irs-2, Homer1, GPRC5B, Grasp). The first gene induction phase occurred at 0-3 h of reperfusion, and the second at 9-15 h, and was validated by in situ hybridization. Four gene clusters displayed a progressive increase in expression over time and included 50 genes linked to cell motility, lipid synthesis and trafficking (i.e. ApoD, NPC1, G3P-dehydrogenase1, and Choline kinase) or cell death-regulating genes such as mitochondrial CLIC. We conclude that a biphasic transcriptional up-regulation of the brain-derived neurotrophic factor (BDNF)-G-protein coupled receptor (GPCR)-mitogen-activated protein (MAP) kinase signaling pathways occurs in surviving tissue, concomitant with a progressive and persistent activation of cell proliferation signifying tissue regeneration, which provide the means for cell survival and postischemic brain plasticity.
Collapse
Affiliation(s)
- Mattias Rickhag
- Laboratory for Experimental Brain Research, Wallenberg Neuroscience Center, University of Lund, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
264
|
Chen C, Bazan NG. Lipid signaling: sleep, synaptic plasticity, and neuroprotection. Prostaglandins Other Lipid Mediat 2005; 77:65-76. [PMID: 16099392 DOI: 10.1016/j.prostaglandins.2005.07.001] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2004] [Accepted: 07/02/2005] [Indexed: 01/25/2023]
Abstract
Increasing evidence indicates that bioactive lipids participate in the regulation of synaptic function and dysfunction. We have demonstrated that signaling mediated by platelet-activating factor (PAF) and cyclooxygenase (COX)-2-synthesized PGE2 is involved in synaptic plasticity, memory, and neuronal protection [Clark GD, Happel LT, Zorumski CF, Bazan NG. Enhancement of hippocampal excitatory synaptic transmission by platelet-activating factor. Neuron 1992; 9:1211; Kato K, Clark GD, Bazan NG, Zorumski CF. Platelet-activating factor as a potential retrograde messenger in CA1 hippocampal long-term potentiation. Nature 1994; 367:175; Izquierdo I, Fin C, Schmitz PK, et al. Memory enhancement by intrahippocampal, intraamygdala or intraentorhinal infusion of platelet-activating factor measured in an inhibitory avoidance. Proc Natl Acad Sci USA 1995; 92:5047; Chen C, Magee CJ, Bazan NG. Cyclooxygenase-2 regulates prostaglandin E2 signaling in hippocampal long-term synaptic plasticity. J Neurophysiol 2002; 87:2851]. Recently, we found that prolonged continuous wakefulness (primarily rapid eye movement (REM)-sleep deprivation, SD) causes impairments in hippocampal long-term synaptic plasticity and hippocampus-dependent memory formation [McDermott CM, LaHoste GJ, Chen C, Musto A, Bazan NG, Magee JC. Sleep deprivation causes behavioral, synaptic, and membrane excitability alterations in hippocampal neurons. J Neurosci 2003; 23:9687]. To explore the mechanisms underlying SD-induced impairments, we have studied several bioactive lipids in the hippocampus following SD. It appears that SD causes increases in prostaglandin D2 (PGD2) and 2-arachidonylglycerol (2-AG), and a decrease in PGE2, suggesting that these lipid messengers participate in memory consolidation during REM sleep. We have also explored the formation of endogenous neuroprotective lipids. Toward this aim, we have used ischemia-reperfusion damage and LC-PDA-ESI-MS-MS-based lipidomic analysis and identified docosanoids derived from synaptic phospholipid-enriched docosahexaenoic acid. Some of the docosanoids exert potent neuroprotective bioactivity [Marcheselli VL, Hong S, Lukiw WJ, et al. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. J Biol Chem 2003; 278:43807; Mukherjee PK, Marcheselli VL, Serhan CN, Bazan, NG. Neuroprotectin D1: A docosahexaenoic acid-derived docosatriene protects human retinal pigment epithelial cells from oxidative stress. Proc Nat Acad Sci USA 2004; 101:8491). Taken together, these observations that signaling lipids participate in synaptic plasticity, cognition, and survival indicate that lipid signaling is closely associated with several functions (e.g; learning and memory, sleep, and experimental stroke) and pathologic events. Alterations in endogenous signaling lipids or their receptors resulting from drug abuse lead to changes in synaptic circuitry and induce profound effects on these important functions. In the present article, we will briefly review bioactive lipids involved in sleep, synaptic transmission and plasticity, and neuroprotection, focusing mainly on our experimental studies and how these signaling molecules are related to functions and implicated in some neurologic disorders.
Collapse
Affiliation(s)
- Chu Chen
- Neuroscience Center of Excellence and Department of Ophthamology, Louisiana State University Health Sciences Center, School of Medicine in New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA 70112, USA
| | | |
Collapse
|
265
|
Riedel M, Strassnig M, Schwarz MJ, Müller N. COX-2 inhibitors as adjunctive therapy in schizophrenia: rationale for use and evidence to date. CNS Drugs 2005; 19:805-19. [PMID: 16185091 DOI: 10.2165/00023210-200519100-00001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A better understanding of the human immune system and its complex interactions has resulted in new insights into the pathoaetiological mechanisms of psychiatric disorders. As a result, new treatment options are being explored. Several findings suggest that an imbalanced immune response is involved in the pathophysiology of schizophrenia. COX-2 inhibitors are known to influence the immune system in a way that may redirect this imbalance. Based on these suggestions, the COX-2 inhibitor celecoxib has been tested as a possible adjunctive therapeutic approach in the treatment of schizophrenia. While the first trial using celecoxib as add-on therapy to an atypical antipsychotic showed a significant beneficial effect, recent studies demonstrated that this effect may be limited to patients with recent-onset schizophrenia.
Collapse
Affiliation(s)
- Michael Riedel
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilan University of Munich, Munich, Germany
| | | | | | | |
Collapse
|
266
|
Dhir A, Naidu PS, Kulkarni SK. Protective effect of cyclooxygenase-2 (COX-2) inhibitors but not non-selective cyclooxygenase (COX)-inhibitors on ethanol withdrawal-induced behavioural changes. Addict Biol 2005; 10:329-35. [PMID: 16318954 DOI: 10.1080/13556210500352964] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Cyclooxygenase (COX) is reported to play a significant role in neurodegeneration. Recent studies have shown that chronic ethanol administration up-regulates cyclooxygenase expression. In the present study we examined the effect of nimesulide (a preferential COX-2 inhibitor), rofecoxib (a highly selective COX-2 inhibitor) or naproxen (a non-selective COX-inhibitor displaying high affinity towards the COX-1 isoenzyme) on alcohol-induced withdrawal symptoms. Mice were made physically dependent on alcohol by the chronic administration of ethanol (2 g/kg of 10% v/v), intragastrically, twice on day 1 and then once-daily on successive days for a total of 7 days. Nimesulide [2.5 mg/kg, intraperitoneally (i.p.)], rofecoxib (2 mg/kg, i.p.) or naproxen (7 mg/kg, i.p.) were administered daily for 7 days before administering alcohol intragastrically. After 24 hours of the last alcohol administration, the treatments were reversed and the mice were tested for withdrawal, so that the animals that had received COX-inhibitors followed 30 minutes later by ethanol on days 1-7 were challenged with saline. Similarly, the animals which received saline followed 30 minutes later by ethanol received only saline. Behavioural analysis revealed hyperlocomotor activity, increased anxious response and increased hyperalgesia in mice. Also, alcohol withdrawal decreased the threshold for Pentylenetetrazole-(PTZ)-induced convulsions. Pretreatment with COX-inhibitors rofecoxib (2 mg/kg, i.p.) or nimesulide (2.5 mg/kg, i.p.) displayed significant protection against ethanol-induced withdrawal symptoms, while naproxen (7 mg/kg, i.p.) was not effective in reversing alcohol-induced withdrawal symptoms. The results of the present study suggest strongly the possible role of cyclooxygenases, particularly COX-2 inhibitors, on ethanol-induced withdrawal symptoms and the potential use of COX-2 inhibitors in their prevention and treatment.
Collapse
Affiliation(s)
- Ashish Dhir
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | | | | |
Collapse
|
267
|
Abdullah L, Ait-Ghezala G, Crawford F, Crowell TA, Barker WW, Duara R, Mullan M. The cyclooxygenase 2 -765 C promoter allele is a protective factor for Alzheimer's disease. Neurosci Lett 2005; 395:240-3. [PMID: 16309832 DOI: 10.1016/j.neulet.2005.10.090] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Revised: 10/23/2005] [Accepted: 10/31/2005] [Indexed: 11/23/2022]
Abstract
The cyclooxygenase-2 enzyme (COX-2) is of particular importance in the inflammatory response and recent findings have demonstrated a considerable role in Alzheimer's disease (AD) pathogenesis. In order to assess the possible putative role of a COX-2 polymorphism (765G/C) in AD, we examined its distribution in 161 community-based controls and 168 AD clinic-based cases previously recruited from memory disorder clinics in Tampa and Miami, Florida. There were no significant differences between the two groups in age/age of onset or gender. A significant difference was observed in the distribution of the COX-2 -765 alleles between AD cases and controls (chi(2) = 6.565, p = .010; OR = .596; CI = [.401-.888], p = .011), with the frequency of the C allele being higher in controls. In addition, a significant difference was observed for this polymorphism by genotype (chi(2) = 6.561, p = .038) and by presence or absence of C+ genotypes (chi(2) = 6.207, p = .013; OR = .464, CI = [.351-.885], p = .013). In this sample, the C allele of COX-2 -765 promoter polymorphism is associated with decreased risk of Alzheimer's disease, a finding which further supports the involvement of COX-2 in AD etiology.
Collapse
Affiliation(s)
- Laila Abdullah
- Roskamp Institute, 2040 Whitfield Ave. Sarasota, FL, USA.
| | | | | | | | | | | | | |
Collapse
|
268
|
Heinrich UR, Brieger J, Selivanova O, Feltens R, Eimermacher A, Schäfer D, Mann WJ. COX-2 expression in the guinea pig cochlea is partly altered by moderate sound exposure. Neurosci Lett 2005; 394:121-6. [PMID: 16289316 DOI: 10.1016/j.neulet.2005.10.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Revised: 09/27/2005] [Accepted: 10/05/2005] [Indexed: 11/16/2022]
Abstract
The cyclooxygenase-2 isoform (COX-2) was found recently to be constitutively expressed in the guinea pig inner ear. To gain knowledge about its role in sound perception, alterations in the COX-2 level of moderate noise-stimulated cochleae were determined. Staining intensities were quantified in different regions using an immunohistochemical staining procedure and computer-assisted system. After 70 dB and 90 dB noise exposure for 1 h at 8000 Hz, COX-2 downregulation was observed in the organ of Corti, which was most prominent in Deiters' cells near Hensen cells and outer hair cells. In pillar cells, COX-2 levels were only slightly reduced after 70 dB but strongly diminished after 90 dB exposure. In Hensen cells, COX-2 was downregulated after 70 dB stimulation, revealing a decreasing COX-2 content from the third to the first turn of the cochlea and a homogeneously reduced enzyme expression in all three turns after 90 dB. The COX-2 content in inner hair cells was nearly identical to unexposed cochleae after 70 dB exposure but significantly reduced after 90 dB stimulation. In spiral ganglion cells, stria vascularis, spiral ligament and limbus, COX-2 expression was unchanged after 70 dB and 90 dB. We suggest that alterations in COX-2 expression might contribute to diminished sensitivity at the cochlea after noise exposure to reduce subsequent noise distress, termed sound conditioning.
Collapse
Affiliation(s)
- Ulf-Rüdiger Heinrich
- Department of Otolaryngology - Head and Neck Surgery, Johannes Gutenberg University Medical School, Mainz, Germany.
| | | | | | | | | | | | | |
Collapse
|
269
|
Yuhas Y, Weizman A, Vanichkin A, Ashkenazi S. Involvement of prostaglandins in an animal model of Shigella-related seizures. J Neuroimmunol 2005; 168:34-9. [PMID: 16139898 DOI: 10.1016/j.jneuroim.2005.06.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Accepted: 06/17/2005] [Indexed: 10/25/2022]
Abstract
We investigated whether prostaglandins (PGs), proinflammatory mediators implicated in excitatory activity, are involved in Shigella-related seizures. Pretreatment with S. dysenteriae sonicate (2LD(50)) enhanced mice response to pentylenetetrazole-induced seizures, without increase of brain concentrations of PGE(2), PGD(2) or PGF(2alpha). Preinjection of NS-398, an inhibitor of cyclooxygenase-2, before treatment with Shigella sonicate, had no effect on seizures. The anticonvulsive PGD(2) increased after injection of 8 LD(50) of Shigella sonicate, which did not enhance seizures (32 pg/mg vs 26 pg/ml, p=0.0063). The findings indicate that PGs are not involved in the enhanced seizure response after exposure to Shigella. However, induction of PGD(2) may play an inhibitory role.
Collapse
Affiliation(s)
- Yael Yuhas
- Laboratory of Infectious Diseases, Felsenstein Medical Research Center, Beilinson Campus, Petach Tikva 49100, Tel Aviv, Israel.
| | | | | | | |
Collapse
|
270
|
Slanina KA, Schweitzer P. Inhibition of cyclooxygenase-2 elicits a CB1-mediated decrease of excitatory transmission in rat CA1 hippocampus. Neuropharmacology 2005; 49:653-9. [PMID: 15936781 DOI: 10.1016/j.neuropharm.2005.04.019] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Revised: 04/19/2005] [Accepted: 04/21/2005] [Indexed: 10/25/2022]
Abstract
Cannabinoid receptor (CB1) ligands decrease excitatory and inhibitory transmission in the hippocampus, but the influence of endogenously formed cannabinoids (eCBs) on basal excitatory transmission remains uncertain. Here, we investigated the influence of eCBs on synaptic transmission in CA1 hippocampus using the slice preparation. Blockade of CB1 with the selective receptor antagonists SR141716 (rimonabant) or AM251 augmented synaptic responses evoked upon stimulation of the Schaffer collaterals. This effect persisted in the presence of bicuculline or CGP55845 to block GABA(A) or GABA(B) receptors, revealing a tonic eCB influence on excitatory transmission. Selective inhibition of cyclooxygenase-2 (COX-2) with meloxicam or NS-398 decreased excitatory responses partly in a CB1-dependent manner, independently of GABA(A) transmission. Paired-pulse paradigms suggested a presynaptic CB1 mechanism to decrease glutamate release. Inhibition of COX-1 or other routes of eCB degradation did not affect synaptic transmission. We conclude that COX-2 regulates the formation of CB1 ligands that decrease hippocampal excitatory transmission.
Collapse
Affiliation(s)
- Kristen A Slanina
- Department of Neuropharmacology, CVN 12, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
271
|
Yin LL, Zhang WY, Li MH, Shen JK, Zhu XZ. CC 05, a novel anti-inflammatory compound, exerts its effect by inhibition of cyclooxygenase-2 activity. Eur J Pharmacol 2005; 520:172-8. [PMID: 16168408 DOI: 10.1016/j.ejphar.2005.06.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 06/14/2005] [Accepted: 06/20/2005] [Indexed: 10/25/2022]
Abstract
In the present study, we examined the anti-inflammation of a novel compound, 4-[5-(3-amino-4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]benzenesulfonamide (CC 05) in vitro and in vivo. In an in vitro cell-based assay, CC 05 inhibited cyclooxygenase (COX)-2-derived prostaglandin E(2) (PGE(2)) synthesis with an IC(50) value of 0.328+/-0.04 microM compared with an IC(50) value of 14.34+/-0.05 microM for the inhibition of COX-1-derived PGE(2) synthesis. In two in vivo rodent models, CC 05 (12.5, 25 and 50 mg/kg, i.g.) is a moderate potential and selective inhibitor of COX-2. It can reduce carrageenan-induced paw edema and PGE(2) production in the inflamed pouch dose-dependently without affecting the PGE(2) production in stomachs. Furthermore, CC 05 had no effect on COX-2 mRNA and protein expression in phorbol 12-myristate 13-acetate (PMA)-differentiated THP-1 human macrophages stimulated with lipopolysaccharide (LPS). These results demonstrate that CC 05 is a novel COX-2 inhibitor and the anti-inflammatory action is not directed towards the transcription or translation of the COX-2 gene but only to the enzymatic activity of the protein.
Collapse
Affiliation(s)
- Lin-Lin Yin
- Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 201203, China
| | | | | | | | | |
Collapse
|
272
|
Bergh MS, Budsberg SC. The Coxib NSAIDs: Potential Clinical and Pharmacologic Importance in Veterinary Medicine. J Vet Intern Med 2005. [DOI: 10.1111/j.1939-1676.2005.tb02741.x] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
273
|
Mraovitch S, Calando Y, Régnier A, Lamproglou I, Vicaut E. Post-seizures amygdaloallocortical microvascular lesion leading to atrophy and memory impairment. Neurobiol Dis 2005; 19:479-89. [PMID: 16023590 DOI: 10.1016/j.nbd.2005.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 01/25/2005] [Accepted: 01/28/2005] [Indexed: 12/01/2022] Open
Abstract
Although the incidence of seizures after a cerebrovascular event including intracerebral hemorrhage has been widely recognized, the present studies have demonstrated that generalized convulsive seizures can cause multifocal amygdaloallocortical hemorrhage and tissue necrosis, the origin of which remains to be established. The seizure-elicited amygdaloallocortical injured area, which we refer to as a focal injury-prone area (FIPA), was caused by cholinergic stimulation of the ventroposterolateral and thalamic reticular nuclei. The amygdaloallocortical injury was preceded by focal absence of neuronal COX-2 and presence of microvascular immunoreactivity to the pro-inflammatory cytokines, IL-1beta and TNF-alpha. The microvascular inflammation was followed by edema and multifocal amygdaloallocortical microhemorrhages, leading to atrophy and cognitive impairment. On the basis of the present results, we conclude that generalized convulsive seizures may be at the origin of amygdaloallocortical microvascular injury suggesting that, in addition to anticonvulsant treatment, an appropriate clinical evaluation and therapy for seizures-associated cerebrovascular accidents should be considered.
Collapse
Affiliation(s)
- Sima Mraovitch
- Laboratoire de Recherche Cérébrovasculaires CNRS URA 641, Université Paris VII, 10, av de Verdun, 75010 Paris, France.
| | | | | | | | | |
Collapse
|
274
|
Steiner AA, Rudaya AY, Robbins JR, Dragic AS, Langenbach R, Romanovsky AA. Expanding the febrigenic role of cyclooxygenase-2 to the previously overlooked responses. Am J Physiol Regul Integr Comp Physiol 2005; 289:R1253-7. [PMID: 16081878 DOI: 10.1152/ajpregu.00371.2005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies on the role of cyclooxygenase (COX)-1 and -2 in fever induced by intravenous LPS have failed to investigate the role of these isoenzymes in the earliest responses: monophasic fever (response to a low, near-threshold dose of LPS) and the first phase of polyphasic fever (response to higher doses). We studied these responses in 96 mice that were COX-1 or COX-2 deficient (-/-) or sufficient (+/+). Each mouse was implanted with a temperature telemetry probe into the peritoneal cavity and a jugular catheter. The study was conducted at a tightly controlled, neutral ambient temperature (31 degrees C). To avoid stress hyperthermia (which masks the onset of fever), all injections were performed through a catheter extension. The +/+ mice responded to intravenous saline with no change in deep body temperature. To a low dose of LPS (1 microg/kg iv), they responded with a monophasic fever. To a higher dose (56 microg/kg), they responded with a polyphasic fever. Neither monophasic fever nor the first phase of polyphasic fever was attenuated in the COX-1 -/- mice, but both responses were absent in the COX-2 -/- mice. The second and third phases of polyphasic fever were also missing in the COX-2 -/- mice. The present study identifies a new, critical role for COX-2 in the mediation of the earliest responses to intravenous LPS: monophasic fever and the first phase of polyphasic fever. It also suggests that no product of the COX-1 gene, including the splice variant COX-1b (COX-3), is essential for these responses.
Collapse
Affiliation(s)
- Alexandre A Steiner
- Systemic Inflammation Laboratory, Trauma Research, St. Joseph's Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, Arizona 85013, USA
| | | | | | | | | | | |
Collapse
|
275
|
Sharifzadeh M, Naghdi N, Khosrovani S, Ostad SN, Sharifzadeh K, Roghani A. Post-training intrahippocampal infusion of the COX-2 inhibitor celecoxib impaired spatial memory retention in rats. Eur J Pharmacol 2005; 511:159-66. [PMID: 15792784 DOI: 10.1016/j.ejphar.2005.01.041] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Revised: 01/10/2005] [Accepted: 01/28/2005] [Indexed: 11/25/2022]
Abstract
In this study, we investigated the effects of intrahippocampal infusion of indomethacin as a non-selective cyclooxygenase inhibitor and celecoxib as a selective cyclooxygenase-2 inhibitor on spatial memory in the Morris water maze. Rats were trained for 3 days; each day included two blocks, and each block contained 4 trials. Tests were performed 48 h after surgery. Bilateral intrahippocampal infusion of indomethacin (0.01, 0.1, or 1 M) did not show any significant effect on spatial memory retention at these concentrations in rats. We also examined effects of infusion of celecoxib (0.02, 0.06, or 0.1 M) on memory retention. Bilateral infusion of 0.1 M celecoxib significantly altered escape latency and traveled distance in rats. These results strongly suggest that cyclooxygenase-2 is involved in spatial memory retention.
Collapse
Affiliation(s)
- Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
276
|
Gendron TF, Brunette E, Tauskela JS, Morley P. The dual role of prostaglandin E2 in excitotoxicity and preconditioning-induced neuroprotection. Eur J Pharmacol 2005; 517:17-27. [PMID: 15964567 DOI: 10.1016/j.ejphar.2005.05.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Revised: 05/20/2005] [Accepted: 05/24/2005] [Indexed: 01/11/2023]
Abstract
Cyclooxygenase-2 is harmful in models of cerebral ischemia yet plays a protective role in preconditioning-induced ischemic tolerance in the heart. This study examined the mechanisms underlying cyclooxygenase-2-mediated neurotoxicity and preconditioning-induced neuroprotection in an in vitro model of cerebral ischemia. Inhibition of cyclooxygenase-2 protects cortical neuronal cultures from death induced by oxygen-glucose deprivation and reduces oxygen-glucose deprivation-induced increases in intracellular Ca(2+) ([Ca(2+)](i)). In the present study, we determined if prostaglandin E(2) (PGE(2)) is responsible for this cyclooxygenase-2-mediated effect. Rat cortical cultures expressed mRNA for the prostanoid EP(1)-EP(4) receptors. PGE(2) reversed the attenuation in [Ca(2+)](i) and the protection offered by cyclooxygenase-2 inhibition during oxygen-glucose deprivation. These effects likely occur via activation of the prostanoid EP(1) receptor since blocking this receptor during oxygen-glucose deprivation reduced [Ca(2+)](i) and neurotoxicity. Next, we considered if the moderate activation of this pathway, by preconditioning cultures with sub-lethal oxygen-glucose deprivation, influenced the development of tolerance to an otherwise lethal oxygen-glucose deprivation insult, 48 h later. Inhibition of cyclooxygenase-2 during oxygen-glucose deprivation-preconditioning abolished preconditioning-induced protection. Furthermore, cultures were rendered tolerant to oxygen-glucose deprivation by the transient exposure to exogenous PGE(2) 24 h prior to the insult, indicating that this product of the cyclooxygenase-2 pathway is sufficient to induce ischemic tolerance. This study shows that cyclooxygenase-2 and PGE(2) are involved in both oxygen-glucose deprivation-induced neurotoxicity and preconditioning-induced neuroprotection. While neurotoxic in the context of lethal oxygen-glucose deprivation, the moderate activation of this signalling pathway confers ischemic tolerance.
Collapse
MESH Headings
- Animals
- Brain
- Calcium/metabolism
- Cell Death/drug effects
- Cell Hypoxia/physiology
- Cells, Cultured
- Cyclooxygenase 2
- Cyclooxygenase 2 Inhibitors
- Cyclooxygenase Inhibitors/pharmacology
- Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology
- Dinoprostone/pharmacology
- Dinoprostone/physiology
- Dose-Response Relationship, Drug
- Female
- Fetus
- Gene Expression/drug effects
- Glucose/deficiency
- Male
- Neuroglia/cytology
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- Pregnancy
- Prostaglandin Antagonists/pharmacology
- Prostaglandin-Endoperoxide Synthases/metabolism
- Pyrazoles/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E/antagonists & inhibitors
- Receptors, Prostaglandin E/genetics
- Receptors, Prostaglandin E, EP1 Subtype
- Receptors, Prostaglandin E, EP4 Subtype
Collapse
Affiliation(s)
- Tania F Gendron
- University of Ottawa, Department of Cellular and Molecular Medicine, Ottawa, Ontario, Canada K1H 8M5.
| | | | | | | |
Collapse
|
277
|
Mollace V, Muscoli C, Masini E, Cuzzocrea S, Salvemini D. Modulation of prostaglandin biosynthesis by nitric oxide and nitric oxide donors. Pharmacol Rev 2005; 57:217-52. [PMID: 15914468 DOI: 10.1124/pr.57.2.1] [Citation(s) in RCA: 251] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The biosynthesis and release of nitric oxide (NO) and prostaglandins (PGs) share a number of similarities. Two major forms of nitric-oxide synthase (NOS) and cyclooxygenase (COX) enzymes have been identified to date. Under normal circumstances, the constitutive isoforms of these enzymes (constitutive NOS and COX-1) are found in virtually all organs. Their presence accounts for the regulation of several important physiological effects (e.g. antiplatelet activity, vasodilation, and cytoprotection). On the other hand, in inflammatory setting, the inducible isoforms of these enzymes (inducible NOS and COX-2) are detected in a variety of cells, resulting in the production of large amounts of proinflammatory and cytotoxic NO and PGs. The release of NO and PGs by the inducible isoforms of NOS and COX has been associated with the pathological roles of these mediators in disease states as evidenced by the use of selective inhibitors. An important link between the NOS and COX pathways was made in 1993 by Salvemini and coworkers when they demonstrated that the enhanced release of PGs, which follows inflammatory mechanisms, was nearly entirely driven by NO. Such studies raised the possibility that COX enzymes represent important endogenous "receptor" targets for modulating the multifaceted roles of NO. Since then, numerous papers have been published extending the observation across various cellular systems and animal models of disease. Furthermore, other studies have highlighted the importance of such interaction in physiology as well as in the mechanism of action of drugs such as organic nitrates. More importantly, mechanistic studies of how NO switches on/off the PG/COX pathway have been undertaken and additional pathways through which NO modulates prostaglandin production unraveled. On the other hand, NO donors conjugated with COX inhibitors have recently found new interest in the understanding of NO/COX reciprocal interaction and potential clinical use. The purpose of this article is to cover the advances which have occurred over the years, and in particular, to summarize experimental data that outline how the discovery that NO modulates prostaglandin production has impacted and extended our understanding of these two systems in physiopathological events.
Collapse
Affiliation(s)
- Vincenzo Mollace
- Faculty of Pharmacy, University of Catanzaro Magna Graecia, Roccelletta di Borgia, Catanazaro, Italy
| | | | | | | | | |
Collapse
|
278
|
Gopez JJ, Yue H, Vasudevan R, Malik AS, Fogelsanger LN, Lewis S, Panikashvili D, Shohami E, Jansen SA, Narayan RK, Strauss KI. Cyclooxygenase-2-specific inhibitor improves functional outcomes, provides neuroprotection, and reduces inflammation in a rat model of traumatic brain injury. Neurosurgery 2005; 56:590-604. [PMID: 15730585 PMCID: PMC1513642 DOI: 10.1227/01.neu.0000154060.14900.8f] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2004] [Accepted: 12/13/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Increases in brain cyclooxygenase-2 (COX2) are associated with the central inflammatory response and with delayed neuronal death, events that cause secondary insults after traumatic brain injury. A growing literature supports the benefit of COX2-specific inhibitors in treating brain injuries. METHODS DFU [5,5-dimethyl-3(3-fluorophenyl)-4(4-methylsulfonyl)phenyl-2(5)H)-furanone] is a third-generation, highly specific COX2 enzyme inhibitor. DFU treatments (1 or 10 mg/kg intraperitoneally, twice daily for 3 d) were initiated either before or after traumatic brain injury in a lateral cortical contusion rat model. RESULTS DFU treatments initiated 10 minutes before injury or up to 6 hours after injury enhanced functional recovery at 3 days compared with vehicle-treated controls. Significant improvements in neurological reflexes and memory were observed. DFU initiated 10 minutes before injury improved histopathology and altered eicosanoid profiles in the brain. DFU 1 mg/kg reduced the rise in prostaglandin E2 in the brain at 24 hours after injury. DFU 10 mg/kg attenuated injury-induced COX2 immunoreactivity in the cortex (24 and 72 h) and hippocampus (6 and 72 h). This treatment also decreased the total number of activated caspase-3-immunoreactive cells in the injured cortex and hippocampus, significantly reducing the number of activated caspase-3-immunoreactive neurons at 72 hours after injury. DFU 1 mg/kg amplified potentially anti-inflammatory epoxyeicosatrienoic acid levels by more than fourfold in the injured brain. DFU 10 mg/kg protected the levels of 2-arachidonoyl glycerol, a neuroprotective endocannabinoid, in the injured brain. CONCLUSION These improvements, particularly when treatment began up to 6 hours after injury, suggest exciting neuroprotective potential for COX2 inhibitors in the treatment of traumatic brain injury and support the consideration of Phase I/II clinical trials.
Collapse
Affiliation(s)
- Jonas J. Gopez
- Department of Neurosurgery, Temple University, School of Medicine, Philadelphia, Pennsylvania
| | - Hongfei Yue
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania
| | - Ram Vasudevan
- Department of Neurosurgery, Temple University, School of Medicine, Philadelphia, Pennsylvania
| | - Amir S. Malik
- Department of Neurosurgery, University of Texas, Houston Medical Center, Houston, Texas
| | - Lester N. Fogelsanger
- Department of Neurosurgery, Temple University, School of Medicine, Philadelphia, Pennsylvania
| | - Shawn Lewis
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Esther Shohami
- Department of Pharmacology, Hebrew University, Jerusalem, Israel
| | - Susan A. Jansen
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania
| | - Raj K. Narayan
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kenneth I. Strauss
- Reprint requests: Kenneth I. Strauss, Ph.D., Department of Neurosurgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML515, Cincinnati, OH 45267-0515.
| |
Collapse
|
279
|
Müller N, Riedel M, Schwarz MJ, Engel RR. Clinical effects of COX-2 inhibitors on cognition in schizophrenia. Eur Arch Psychiatry Clin Neurosci 2005; 255:149-51. [PMID: 15549344 DOI: 10.1007/s00406-004-0548-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Accepted: 09/03/2004] [Indexed: 10/26/2022]
Abstract
An activation of pro-inflammatory cytokines in the central nervous system is associated with cognitive disturbances. This process is mediated by prostaglandins and cyclo-oxygenase-2 (COX-2). COX-2 inhibitors have been suggested to show beneficial effects in disorders associated with cognitive disturbance, although clinical effects on cognition have not been shown until today. Data from a schizophrenia study were reevaluated under the aspect whether an effect on the positive and negative syndrome scale (PANSS) factor cognition can be observed during therapy with the COX-2 inhibitor celecoxib add on to risperidone in comparison to riperidone alone. Beside the effect on the PANSS total score, the effect on the cognition factor was the most pronounced using the analysis of covariance compared to all other factors of the PANSS (p < 0.06). Although suggestions of basic research led to an expected therapeutic effect of COX-2 inhibitors on cognition, this effect could not yet be shown clinically. In schizophrenia, the effect on cognition contributes to the therapeutic effect of COX-2 inhibitors.
Collapse
Affiliation(s)
- Norbert Müller
- Hospital for Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität, Nussbaumstr. 7, 80336 München, Germany.
| | | | | | | |
Collapse
|
280
|
Li Z, Jansen M, Ogburn K, Salvatierra L, Hunter L, Mathew S, Figueiredo-Pereira ME. Neurotoxic prostaglandin J2 enhances cyclooxygenase-2 expression in neuronal cells through the p38MAPK pathway: a death wish? J Neurosci Res 2005; 78:824-36. [PMID: 15523637 DOI: 10.1002/jnr.20346] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The role of the proinflammatory and inducible form of cyclooxygenases (COX-2) in neurodegeneration is not well defined. Some of its metabolic products, such as prostaglandins (PG) of the J2 series, are known to be neurotoxic. Here we demonstrate that PGJ2 enhances COX-2 gene expression without elevating COX-1 levels in neuronal cells. PGJ2 also increased PGE2 production, establishing that the de novo synthesized COX-2 is enzymatically active. PGJ2 derivatives, such as 15d-PGJ2, are known activators of PPARgamma, a nuclear receptor that activates gene expression. However, the selective PPARgamma agonist ciglitazone failed to up-regulate COX-2, indicating that the PGJ2 effect on COX-2 is PPARgamma independent. Furthermore, PGJ2 stabilized IkappaBalpha levels, indicating that NFkappaB is not active under these conditions. The blocking of neuronal NFkappaB activity by PGJ2 may be an important contributor to its neurotoxicity, insofar as NFkappaB transactivation seems to be required for neuronal survival in the CNS. Interleukin-1 (IL1) is a proinflammatory cytokine known to stimulate the expression of genes associated with inflammation, including COX-2. Notably, IL1 mRNA levels in the neuronal cells were increased by PGJ2 treatment. The proinflammatory cytokine may mediate COX-2 up-regulation by PGJ2 through p38MAPK and not JNK activation, in that only an inhibitor of the former prevented the COX-2 increase. Thiol-reducing agents, such as N-acetylcysteine, protected the neuronal cells from the deleterious effects of PGJ2, whereas ascorbic acid did not. Collectively, our findings suggest that proinflammatory conditions that lead to COX-2 up-regulation and the concomitant production of PGJ2 initiate a mechanism of self-destruction through an autotoxic loop between PGJ2 and COX-2 that may exacerbate neurodegeneration beyond a point of no return. Thiol-reducing antioxidants may offer an optimal strategy for halting this neurodegenerative process.
Collapse
Affiliation(s)
- Zongmin Li
- Department of Biological Sciences, Hunter College of City University of New York, New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
281
|
Reid MS, Angrist B, Baker S, Woo C, Schwartz M, Montgomery A, Majewska D, Robinson J, Rotrosen J. A placebo-controlled screening trial of celecoxib for the treatment of cocaine dependence. Addiction 2005; 100 Suppl 1:32-42. [PMID: 15730348 DOI: 10.1111/j.1360-0443.2005.00989.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS To conduct a medication screening trial study on the efficacy of celecoxib versus placebo for the treatment of cocaine dependence. DESIGN A modified blinded, parallel group study in an outpatient setting using the Cocaine Rapid Efficacy and Safety Trials (CREST) study design. SETTING The study was performed at the New York Medications Development Research Unit (MDRU). PARTICIPANTS All participants met Diagnostic and Statistical Manual version IV (DSM-IV) criteria for cocaine dependence and provided at least two urine samples positive for benzoylecgonine (BE) during the 2-week screening period. Twenty-three participants were enrolled in the treatment phase of the study. INTERVENTION After a 2-week screening period, subjects were assigned randomly to receive either celebrex (200 mg/day) or placebo for an 8-week treatment period. All subjects also received individual cognitive behavioral counseling during treatment. MEASUREMENTS Primary outcome measures included quantitative urine benzoylecgonine (BE) levels, self-report of drug use and global impression scores. Secondary outcomes included cocaine craving, study retention and related psychosocial measures. Safety measures included adverse event monitoring, vital signs and extrapyramidal side-effects tests. RESULTS Study retention was similar across both treatment groups and safety measures indicated that celecoxib was moderately tolerated. Cocaine use, as measured by self-report and urine BE levels at end of treatment, indicated weaker improvement in the celecoxib group. Reductions in the intensity of cocaine craving were also weaker in the celecoxib group. Cocaine abstinence rates, global impression scores and all other related psychometric measures did not differ significantly between treatment groups. CONCLUSION This study does not support the effectiveness of celecoxib for the treatment of cocaine dependence.
Collapse
Affiliation(s)
- Malcolm S Reid
- Department of Psychiatry, New York University School of Medicine, VA New York Harbor Healthcare System, New York, NY 10010, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
282
|
Müller N, Strassnig M, Schwarz MJ, Ulmschneider M, Riedel M. COX-2 inhibitors as adjunctive therapy in schizophrenia. Expert Opin Investig Drugs 2005; 13:1033-44. [PMID: 15268640 DOI: 10.1517/13543784.13.8.1033] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cyclooxygenase-2 (COX-2) is constitutively expressed in the central nervous system, and is thought to have an important functional role therein. COX-2 interacts with neurotransmitters such as acetylcholine, 5-hydroxytryptamine and glutamate but is also involved in the regulation of the central nervous system immune system and in inflammation via the effects of prostaglandins, in particular prostaglandin E2. A general therapeutic effect of the COX-2 inhibitor celecoxib on symptoms of schizophrenia was observed during a prospective, randomised, double-blind study of celecoxib add-on treatment to the atypical antipsychotic risperidone. The results from this trial of adjunctive therapy with a COX-2 inhibitor in schizophrenia are encouraging, and the findings support the view that an immunological/inflammatory process is involved in the pathogenesis of the disease. The add-on to an antipsychotic design of the study was chosen due to ethical reasons; in less acute schizophrenic states a monotherapy with COX-2 inhibitors would be interesting. From a theoretical point of view, other psychiatric indications for selective COX-2 inhibitors are discussed. COX-2 inhibitors have failed to show therapeutic effects in Alzheimer's disease but studies from basic research and a clinical perspective suggest it has an effect on disturbed cognition. In depression, however, signs of inflammation have been described for many years. Although results of clinical studies with COX-2 inhibitors in depression are still lacking, clinical improvement of a depressive syndrome has been observed in patients who have been treated with the COX-2 inhibitor rofecoxib due to other indications. These preliminary clinical data are encouraging for clinical therapeutic effects of the selective COX-2 inhibitors in psychiatric disorders, although these effects have to be confirmed in larger clinical studies.
Collapse
Affiliation(s)
- Norbert Müller
- Ludwig-Maximilans University, Department of Psychiatry and Psychotherapy, Nussbaumstrasse 7, D-80336 Munich, Germany.
| | | | | | | | | |
Collapse
|
283
|
Engelberg H. Pathogenic factors in vascular dementia and Alzheimer's disease. Multiple actions of heparin that probably are beneficial. Dement Geriatr Cogn Disord 2005; 18:278-98. [PMID: 15286460 DOI: 10.1159/000080034] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/04/2004] [Indexed: 12/22/2022] Open
Abstract
The following areas are discussed in this review: atherogenesis; cerebrovascular factors; hypoperfusion; beta-amyloid production; beta-amyloid fibril formation; beta-sheets; metal cations; reactive oxygen species/free radicals; chronic inflammatory factors; endogenous plasma heparin; lipoprotein lipase; polyamines; protein kinase C; casein kinases; phospholipase A2; serine proteases; myeloperoxidase; cyclooxygenase 2; cysteine proteases; caspases; proprotein convertases; aspartic proteases; cyclin proteinases; thrombin; tau hyperphosphorylation; advanced glycosylation end products; activator protein 1; calcium; apolipoprotein E epsilon4; histamine; blood-brain barrier; glutamate; transglutaminase; insulin-like growth factor 1.
Collapse
|
284
|
Abstract
A key goal in functional neuroimaging is to use signals that are related to local changes in metabolism and blood flow to track the neuronal correlates of mental activity. Recent findings indicate that the dendritic processing of excitatory synaptic inputs correlates more closely than the generation of spikes with brain imaging signals. The correlation is often nonlinear and context-sensitive, and cannot be generalized for every condition or brain region. The vascular signals are mainly produced by increases in intracellular calcium in neurons and possibly astrocytes, which activate important enzymes that produce vasodilators to generate increments in flow and the positive blood oxygen level dependent signal. Our understanding of the cellular mechanisms of functional imaging signals places constraints on the interpretation of the data.
Collapse
Affiliation(s)
- Martin Lauritzen
- Department of Clinical Neurophysiology, Glostrup Hospital, DK-2600 Glostrup, Denmark.
| |
Collapse
|
285
|
Chen C, Bazan NG. Endogenous PGE2Regulates Membrane Excitability and Synaptic Transmission in Hippocampal CA1 Pyramidal Neurons. J Neurophysiol 2005; 93:929-41. [PMID: 15653788 DOI: 10.1152/jn.00696.2004] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The significance of cyclooxygenases (COXs), the rate-limiting enzymes that convert arachidonic acid (AA) to prostaglandins (PGs) in the brain, is unclear, although they have been implicated in inflammatory responses and in some neurological disorders such as epilepsy and Alzheimer's disease. Recent evidence that COX-2, which is expressed in postsynaptic dendritic spines, regulates PGE2signaling in activity-dependent long-term synaptic plasticity at hippocampal perforant path-dentate granule cell synapses, suggests an important role of the COX-2–generated PGE2in synaptic signaling. However, little is known of how endogenous PGE2regulates neuronal signaling. Here we showed that endogenous PGE2selectively regulates fundamental membrane and synaptic properties in the hippocampus. Somatic and dendritic membrane excitability was significantly reduced when endogenous PGE2was eliminated with a selective COX-2 inhibitor in hippocampal CA1 pyramidal neurons in slices. Exogenous application of PGE2produced significant increases in frequency of firing, excitatory postsynaptic potentials (EPSP) amplitude, and temporal summation in slices treated with the COX-2 inhibitor. The PGE2-induced increase in membrane excitability seemed to result from its inhibition of the potassium currents, which in turn, boosted dendritic Ca2+influx during dendritic-depolarizing current injections. In addition, the PGE2-induced enhancement of EPSPs was blocked by eliminating both PKA and PKC activities. These findings indicate that endogenous PGE2dynamically regulates membrane excitability, synaptic transmission, and plasticity and that the PGE2-induced synaptic modulation is mediated via cAMP-PKA and PKC pathways in rat hippocampal CA1 pyramidal neurons.
Collapse
Affiliation(s)
- Chu Chen
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | | |
Collapse
|
286
|
Imai T, Hasumura M, Onose JI, Ueda M, Takizawa T, Cho YM, Hirose M. Development of invasive follicular cell carcinomas in a rat thyroid carcinogenesis model: biological impact of capsular inflammation and reduced cyclooxygenase-2 expression. Cancer Sci 2005; 96:31-7. [PMID: 15649252 PMCID: PMC11159682 DOI: 10.1111/j.1349-7006.2005.00002.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
We have previously reported that thyroid capsular inflammation induced by sulfadimethoxine (SDM), a goitrogen, might play a role in development of invasive follicular cell adenocarcinomas in rats initiated with N-bis(2-hydroxypropyl)nitrosamine (DHPN). The present study was designed to examine the role of cyclooxygenase (COX)-2, widely known to be up-regulated in inflammatory states, during chemically induced rat thyroid carcinogenesis. Male F344 rats received a subcutaneous DHPN (2800 mg/kg) injection, and 1 week later were allowed free access to drinking water containing antithyroidal propylthiouracil (PTU, 0.003%) or SDM (0.1%) for 4 or 10 weeks. Control groups receiving goitrogen alone and no treatment were also included. At week 4, diffuse follicular cell hyperplasia was induced in all PTU- and SDM-treated groups, along with fibrous capsular thickening and capsular thickening with inflammation, respectively. Additionally, multiple focal follicular cell hyperplasias and adenomas were observed in the DHPN + PTU and DHPN + SDM cases. At week 10, adenocarcinomas invasive to the capsule and restricted to the capsular adjacent region, were frequent in the DHPN + SDM group, but not observed in the animals given DHPN + PTU. Western blots and immunohistochemistry revealed constitutive COX-2 expression in non-neoplastic follicular cells of the control and all of the PTU- and SDM-treated rats. However, COX-2 reactivity was significantly reduced or negative in the preneoplastic/neoplastic lesions in the DHPN-treated groups. In fibrous or inflamed thickened capsules, only a few component cells with inflammatory elements were positive for COX-2, and there was no significant difference in this regard between the PTU and SDM treatments. The present results suggest that capsular inflammation could play a role in development of invasive carcinomas, but COX-2 expression does not make a major contribution.
Collapse
Affiliation(s)
- Toshio Imai
- Division of Pathology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-Ku, Tokyo 158-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
287
|
Zhu P, Genc A, Zhang X, Zhang J, Bazan NG, Chen C. Heterogeneous expression and regulation of hippocampal prostaglandin E2 receptors. J Neurosci Res 2005; 81:817-26. [PMID: 16041798 DOI: 10.1002/jnr.20597] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Although prostaglandin E2 (PGE2) has been shown to be critical to hippocampal synaptic signaling and neuronal survival, it is still not clear which subtypes of PGE2 receptors (EPs) are expressed and how these EPs are regulated in the hippocampus. To address these questions, the expression of the EPs was profiled in the hippocampus. Messenger RNAs and proteins of the four receptors, EP 1-4, were detected both in the hippocampus and in the neocortex. EP 2 and EP 3 appeared in greater abundance, whereas EP 1 and EP 4 were barely detectable. EP 1, EP 2 and EP 4 were mainly colocalized with synaptophysin, suggesting the presence of EP 1, EP 2, and EP 4 in presynaptic terminals. It appeared that interleukin-1 beta increased the expression of EP 2 and EP 4 mRNAs. A blockade of synaptic transmission with either tetrodotoxin or MK-801 plus 6,7-dinitroquinoxaline-2,3-dione (DNQX) for 6 hr increased EP 3 and EP 4 mRNA, whereas high K(+) (90 mM) or 4-aminopyridine enhanced EP 2 and EP 4. The EP 1 level did not change significantly under these conditions. The expressions of EP 2, EP 4, and EP 3 were further elevated or reduced in neurons treated with high K(+) for 24 hr. However, mRNA of EP 3 was down-regulated in neurons treated with tetrodotoxin or MK-801 plus DNQX for 24 hr. In addition, both EP 2 and EP 4 mRNAs were up-regulated within 4 hr after high-frequency stimulation associated with long-term potentiation induction in hippocampal slices. Our results indicate that the four EPs are heterogeneously expressed in the hippocampus, and their expression is differentially regulated by neuronal activities, suggesting that EPs may actively participate in hippocampal synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Peimin Zhu
- Neuroscience Center, School of Medicine, LSU Health Sciences Center, New Orleans, Louisiana
| | | | | | | | | | | |
Collapse
|
288
|
Wang H, Hitron IM, Iadecola C, Pickel VM. Synaptic and vascular associations of neurons containing cyclooxygenase-2 and nitric oxide synthase in rat somatosensory cortex. ACTA ACUST UNITED AC 2004; 15:1250-60. [PMID: 15616132 DOI: 10.1093/cercor/bhi008] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Cyclooxygenase-2 (COX-2) is a rate-limiting enzyme for prostanoid synthesis that is present in cortical pyramidal neurons and highly implicated in control of cerebral blood flow during neural activity. We examined the electron microscopic localization of COX-2 and neuronal nitric oxide synthase (nNOS), a functionally related enzyme, in the somatosensory cortex of rat brain to determine the relevant functional sites. COX-2 immunoreactivity was detected in significantly more somatodendritic than axonal profiles, while nNOS was more often seen in axon terminals. The dendritic COX-2 was localized to endomembranes near synaptic inputs from axon terminals, some of which contained nNOS. Conversely, COX-2 terminals formed asymmetric, excitatory-type synapses with dendrites containing nNOS. The dendritic and axonal profiles containing COX-2 as well as those containing nNOS were minimally separated from penetrating arterioles and capillaries by filamentous glial processes. The perivascular COX-2 labeled terminals were among those that also formed axo-dendritic synapses, suggesting that the release of prostanoids and/or excitatory transmitters from a single terminal may simultaneously affect neuronal activity and cerebral blood flow. Thus, COX-2 has a compartmental distribution in somatosensory cortical neurons consistent with the local neuronal synthesis of prostanoids that are involved in neurovascular coupling and whose actions are modulated by nitric oxide.
Collapse
Affiliation(s)
- Hong Wang
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
289
|
Bouras EP, Burton DD, Camilleri M, Stephens DA, Thomforde GM. Effect of cyclooxygenase-2 inhibitors on gastric emptying and small intestinal transit in humans. Neurogastroenterol Motil 2004; 16:729-35. [PMID: 15601422 DOI: 10.1111/j.1365-2982.2004.00547.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Endogenous prostaglandins regulate smooth muscle activity; prostaglandins and cyclooxygenase (COX) inhibitors influence gastrointestinal motility in inflammatory states such as postoperative ileus in animal models. The objective of this study was to evaluate the effects of two COX-2 inhibitors on gastric emptying and intestinal transit in healthy humans. In a double-blind, placebo-controlled, parallel-group study, 66 healthy volunteers were randomized to one of two commercially available oral COX-2 inhibitors (celecoxib and rofecoxib), cisapride (positive control), or placebo. Following 7 days on therapy, study participants underwent a test of gastric emptying and small bowel transit of liquids and solids using scintigraphy. Data were analysed using Kruskal-Wallis (ANOVA on ranks)and Mann-Whitney rank sum tests. There were significant group effects on transit of solids: gastric emptying (ANOVA, P = 0.005) and small bowel transit (ANOVA, P = 0.056). However, neither COX-2 inhibitor significantly accelerated the liquid or solid gastric emptying or small bowel transit compared with placebo. The positive control, cisapride, accelerated gastric emptying of solids (post-lag slope of gastric emptying, P < 0.05), and small bowel transit of solids (t10%, P = 0.016). At maximum clinically approved dosages, celecoxib and rofecoxib have no significant effects on gastric emptying or small intestinal transit in healthy humans. Cisapride accelerates gastric emptying and small bowel transit in healthy humans.
Collapse
Affiliation(s)
- E P Bouras
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER) Program, Gastroenterology Research Unit, Mayo Clinic College of Medicine, Jacksonville, FL, USA.
| | | | | | | | | |
Collapse
|
290
|
Devesa I, Alcaraz MJ, Riguera R, Ferrándiz ML. A new pyrazolo pyrimidine derivative inhibitor of cyclooxygenase-2 with anti-angiogenic activity. Eur J Pharmacol 2004; 488:225-30. [PMID: 15044055 DOI: 10.1016/j.ejphar.2004.02.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2003] [Revised: 02/05/2004] [Accepted: 02/10/2004] [Indexed: 11/25/2022]
Abstract
In a previous study, we reported a new pyrazolo pyrimidine derivative, N(4)-benzyl-N(6),N(6)-dimethyl-1-1(tert-butyl)-1H-pyrazolo[3,4-d]pyrimidine-6,4-diamine (DPP), which inhibited potently cyclooxygenase-2 activity in intact cell assays with minor activity against cyclooxygenase-1 (IC(50)=0.9 nM for cyclooxygenase-2 versus IC(50)=59.6 nM for cyclooxygenase-1). In the present work, this behaviour was confirmed in vivo by using the 24-h zymosan-injected mouse air pouch model (ID(50)=1.36 nM/pouch for prostaglandin E(2) level). We also studied the possible beneficial effect of DPP in the angiogenesis-dependent murine air pouch granuloma and rat paw carrageenan-induced hyperalgesia models. DPP exerted analgesic and anti-angiogenic (52% reduction in angiogenesis at 10 mg/kg, i.p.) effects that may be associated with inhibition of cyclooxygenase-2 activity.
Collapse
Affiliation(s)
- Isabel Devesa
- Departamento de Farmacología, Universidad de Valencia, 46100 Burjasot, Valencia, Spain
| | | | | | | |
Collapse
|
291
|
Vazquez-Tello A, Fan L, Hou X, Joyal JS, Mancini JA, Quiniou C, Clyman RI, Gobeil F, Varma DR, Chemtob S. Intracellular-specific colocalization of prostaglandin E2synthases and cyclooxygenases in the brain. Am J Physiol Regul Integr Comp Physiol 2004; 287:R1155-63. [PMID: 15284079 DOI: 10.1152/ajpregu.00077.2004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prostaglandin E2(PGE2) is the major primary prostaglandin generated by brain cells. However, the coordination and intracellular localization of the cyclooxygenases (COXs) and prostaglandin E synthases (PGESs) that convert arachidonic acid to PGE2in brain tissue are not known. We aimed to determine whether microsomal and cytosolic PGES (mPGES-1 and cPGES) colocalize and coordinate activity with either COX-1 or COX-2 in brain tissue, particularly during development. Importantly, we found that cytosolic PGES also associates with microsomes (cPGES-m) from the cerebrum and cerebral vasculature of the pig and rat as well as microsomes from various cell lines; this seemed dependent on the carboxyl terminal 35-amino acid domain and a cysteine residue (C58) of cPGES. In microsomal membranes from the postnatal brain and cerebral microvessels of mature animals, cPGES-m colocalized with both COX-1 and COX-2, whereas mPGES-1 was undetectable in these microsomes. Accordingly, in this cell compartment, cPGES could coordinate its activity with COX-2 and COX-1 (partly inhibited by NS398); albeit in microsomes of the brain microvasculature from newborns, mPGES-1 was also present. In contrast, in nuclei of brain parenchymal and endothelial cells, mPGES-1 and cPGES colocalized exclusively with COX-2 (determined by immunoblotting and immunohistochemistry); these PGESs contributed to conversion of PGH2into PGE2. Hence, contrary to a previously proposed model of exclusive COX-2/mPGES-1 coordination, COX-2 can coordinate with mPGES-1 and/or cPGES in the brain, depending on the cell compartment and the age group.
Collapse
Affiliation(s)
- Alejandro Vazquez-Tello
- Deptartment of Pediatrics, Research Center of Hôpital Sainte-Justine, 3175, Côte Sainte-Catherine, Montréal, Québec, Canada H3T 1C5
| | | | | | | | | | | | | | | | | | | |
Collapse
|
292
|
|
293
|
Simmons DL, Botting RM, Hla T. Cyclooxygenase isozymes: the biology of prostaglandin synthesis and inhibition. Pharmacol Rev 2004; 56:387-437. [PMID: 15317910 DOI: 10.1124/pr.56.3.3] [Citation(s) in RCA: 1212] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) represent one of the most highly utilized classes of pharmaceutical agents in medicine. All NSAIDs act through inhibiting prostaglandin synthesis, a catalytic activity possessed by two distinct cyclooxygenase (COX) isozymes encoded by separate genes. The discovery of COX-2 launched a new era in NSAID pharmacology, resulting in the synthesis, marketing, and widespread use of COX-2 selective drugs. These pharmaceutical agents have quickly become established as important therapeutic medications with potentially fewer side effects than traditional NSAIDs. Additionally, characterization of the two COX isozymes is allowing the discrimination of the roles each play in physiological processes such as homeostatic maintenance of the gastrointestinal tract, renal function, blood clotting, embryonic implantation, parturition, pain, and fever. Of particular importance has been the investigation of COX-1 and -2 isozymic functions in cancer, dysregulation of inflammation, and Alzheimer's disease. More recently, additional heterogeneity in COX-related proteins has been described, with the finding of variants of COX-1 and COX-2 enzymes. These variants may function in tissue-specific physiological and pathophysiological processes and may represent important new targets for drug therapy.
Collapse
Affiliation(s)
- Daniel L Simmons
- Department of Chemistry and Biochemistry, E280 BNSN, Brigham Young University, Provo, UT 84604, USA.
| | | | | |
Collapse
|
294
|
Docquier MA, Lavand'homme P, Boulanger V, Collet V, De Kock M. Questioning the cardiocirculatory excitatory effects of opioids under volatile anaesthesia. Br J Anaesth 2004; 93:408-13. [PMID: 15220166 DOI: 10.1093/bja/aeh216] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Opioid-induced hyperalgesia has been demonstrated in awake animals. We observed an increased haemodynamic reactivity in response to noxious stimuli in rats under sevoflurane anaesthesia treated with a very low dose of sufentanil. The aim of this investigation was to determine whether the two phenomena share a common origin: an opioid-induced excitatory reaction. To address this, we administered several drugs with proven efficacy in opioid hyperalgesia to rats presenting with haemodynamic hyper-reactivity. METHODS The MACbar of sevoflurane was measured in controls and in animals treated with sufentanil 0.005 micro g kg(-1) min(-1) before and after administration of i.v. (0.25, 0.5 mg kg(-1)) and intrathecal (i.t.) (250 micro g) ketamine, i.v. (0.5, 1 mg kg(-1)) and i.t. (30 micro g) MK-801(NMDA antagonist), i.v. (0.1, 0.5 mg kg(-1)) naloxone, i.v. (10 mg kg(-1)) and i.t. (50, 100 micro g) ketorolac or i.t. (100, 150 micro g) meloxicam (COX-2 inhibitor). RESULTS Sufentanil 0.005 micro g kg(-1) min(-1) significantly increased MACbar (3.2 (sd 0.3) versus 1.9 (0.3) vol%). With the exception of naloxone, all drugs displayed a significant MACbar-sparing effect (>50%) in controls. Naloxone completely prevented haemodynamic hyperactivity. Two patterns of reaction were recorded for the other drugs: either hyper-reactivity was suppressed and the MACbar-sparing effect was maintained (i.t. ketamine, i.t. MK-801, i.t. ketorolac [100 micro g], i.t. meloxicam [150 micro g]) or hyper-reactivity was blocked but MACbar-sparing effect was lost (i.v. ketamine [0.5 mg kg(-1)], i.v. MK-801 [0.5, 1 mg kg(-1)], i.v. ketorolac [10 micro g kg(-1)], i.t. ketorolac [50 micro g], i.t. meloxicam [100 micro g]). CONCLUSIONS We have demonstrated that low-dose sufentanil-induced haemodynamic hyper-reactivity is an excitatory micro -opiate-related phenomenon. This effect is reversed by drugs effective in treating opiate-induced hyperalgesia.
Collapse
Affiliation(s)
- M-A Docquier
- Department of Anaesthesiology, Laboratory of Anaesthesia, University of Louvain, St Luc Hospital, av. Hippocrate 10-1821, 1200 Brussels, Belgium
| | | | | | | | | |
Collapse
|
295
|
Minghetti L. Cyclooxygenase-2 (COX-2) in Inflammatory and Degenerative Brain Diseases. J Neuropathol Exp Neurol 2004; 63:901-10. [PMID: 15453089 DOI: 10.1093/jnen/63.9.901] [Citation(s) in RCA: 594] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cyclooxygenase (COX) catalyses the first committed step in the synthesis of prostanoids, a large family of arachidonic acid metabolites comprising prostaglandins, prostacyclin, and thromboxanes, and is a major target of non-steroidal anti-inflammatory drugs (NSAIDs). COX exists as constitutive and inducible isoforms. COX-2 is the inducible isoform, rapidly expressed in several cell types in response to growth factors, cytokines, and pro-inflammatory molecules. Since its discovery in the early 1990s, COX-2 has emerged as a major player in inflammatory reactions in peripheral tissues. By extension, COX-2 expression in brain has been associated with pro-inflammatory activities, thought to be instrumental in neurodegenerative processes of several acute and chronic diseases. However, 2 major aspects should be borne in mind. First, in the central nervous system, COX-2 is expressed under normal conditions and contributes to fundamental brain functions, such as synaptic activity, memory consolidation, and functional hyperemia. Second, "neuroinflammation" is a much more controlled reaction than inflammation in peripheral tissues, and in many cases is triggered and sustained by activation of resident cells, particularly microglia. In spite of the intense research of the last decade, the evidence of a direct role of COX-2 in neurodegenerative events is still controversial. This article will review new data in this area, focusing on some major human neurological diseases, such as multiple sclerosis, amyotrophic lateral sclerosis, Parkinson disease, Creutzfeldt-Jakob disease, and Alzheimer disease. Furthermore, the emerging role of COX-2 in behavioral and cognitive functions will be discussed.
Collapse
Affiliation(s)
- Luisa Minghetti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
296
|
Zewde T, Mattson DL. Inhibition of cyclooxygenase-2 in the rat renal medulla leads to sodium-sensitive hypertension. Hypertension 2004; 44:424-8. [PMID: 15314032 DOI: 10.1161/01.hyp.0000140924.91479.03] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cyclooxygenase-2 expression in the renal medulla is regulated by dietary salt intake. The present study was performed to determine the influence of chronic inhibition of medullary cyclooxygenase-2 on arterial blood pressure in conscious Sprague-Dawley rats maintained on a high-salt (4% NaCl) or a low-salt (0.4% NaCl) diet. Rats were uninephrectomized and instrumented with femoral arterial and femoral vein or renal medullary interstitial catheters. Each rat received a continuous medullary or intravenous infusion of saline (0.5 mL per hour) for 3 control days, followed by infusion of the cyclooxygenase-2 inhibitor NS-398 (10 mg/kg per day) for 5 days. Medullary interstitial infusion of NS-398 significantly increased mean arterial pressure in the 4% NaCl group from 126+/-2 to 146+/-2 mm Hg (n=6) but did not alter blood pressure in the 0.4% NaCl group (n=6). Intravenous infusion of NS-398 to rats on the 4.0% NaCl diet also failed to alter mean arterial pressure (n=5). To test the blood pressure effect of a mechanistically different inhibitor of cyclooxygenase-2, an antisense oligonucleotide against cyclooxygenase-2 (18-mer; 8 nmol per hour) was infused into the renal medulla of rats maintained on a high-salt diet. Administration of the antisense oligonucleotide reduced cyclooxygenase-2 immunoreactive protein by 36% and significantly increased mean arterial pressure from 127+/-2 to 147+/-2 mm Hg (n=6). Renal medullary interstitial infusion of a scrambled oligonucleotide did not alter arterial pressure (n=5). These results demonstrate the importance of cyclooxygenase-2 in the renal medulla in maintaining blood pressure during high-salt intake.
Collapse
Affiliation(s)
- Tewabech Zewde
- Department of Physiology, Medical College of Wisconsin, Milwaukee 53226, USA
| | | |
Collapse
|
297
|
Pérez González M, Dias LC, Helguera AM, Rodríguez YM, de Oliveira LG, Gomez LT, Diaz HG. TOPS-MODE based QSARs derived from heterogeneous series of compounds. Applications to the design of new anti-inflammatory compounds. Bioorg Med Chem 2004; 12:4467-75. [PMID: 15265497 DOI: 10.1016/j.bmc.2004.05.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2004] [Revised: 05/22/2004] [Accepted: 05/26/2004] [Indexed: 11/30/2022]
Abstract
A new application of TOPological Sub-structural MOlecular DEsign (TOPS-MODE) was carried out in anti-inflammatory compounds using computer-aided molecular design. Two series of compounds, one containing anti-inflammatory and the other containing nonanti-inflammatory compounds were processed by a k-means cluster analysis in order to design the training and prediction sets. A linear classification function to discriminate the anti-inflammatory from the inactive compounds was developed. The model correctly and clearly classified 88% of active and 91% of inactive compounds in the training set. More specifically, the model showed a good global classification of 90%, that is, (399 cases out of 441). While in the prediction set, they showed an overall predictability of 88% and 84% for active and inactive compounds, being the global percentage of good classification of 85%. Furthermore this paper describes a fragment analysis in order to determine the contribution of several fragments towards anti-inflammatory property, also the present of halogens in the selected fragments were analyzed. It seems that the present TOPS-MODE based QSAR is the first alternate general 'in silico' technique to experimentation in anti-inflammatory discovery.
Collapse
Affiliation(s)
- Maykel Pérez González
- Unit of Service, Drug Design Department, Experimental Sugar Cane Station Villa Clara-Cienfuegos, Villa Clara, Ranchuelo 53100, Cuba.
| | | | | | | | | | | | | |
Collapse
|
298
|
Manabe Y, Anrather J, Kawano T, Niwa K, Zhou P, Ross ME, Iadecola C. Prostanoids, not reactive oxygen species, mediate COX-2-dependent neurotoxicity. Ann Neurol 2004; 55:668-75. [PMID: 15122707 DOI: 10.1002/ana.20078] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The prostaglandin synthesizing enzyme cyclooxygenase-2 (COX-2) has emerged as a critical pathogenic factor in brain diseases associated with activation of N-methyl-D-aspartate (NMDA) receptors, including stroke and neurodegenerative diseases. However, the COX-2 reaction products responsible for these deleterious effects have not been identified. In particular, the relative contribution to the neurotoxicity of COX-2-derived prostanoids and reactive oxygen species has not been defined. We found that the brain damage produced by direct injection of NMDA into the somatosensory cortex is attenuated by the COX-2 inhibitor NS-398 or in COX-2-null mice, but that the associated production of free radicals is not. Furthermore, COX-2 inhibition reduces the lesions even if the deleterious effects of free radicals are eliminated by the scavenger superoxide dismutase. The protection exerted by NS-398 is counteracted by a stable analog of prostaglandin E2. The findings directly implicate COX-2-derived prostanoids, rather then radicals, in the COX-2-dependent component of the damage mediated by NMDA receptors and strengthen the rationale for using COX-2 inhibitors in the treatment of neurological diseases associated with glutamate neurotoxicity.
Collapse
Affiliation(s)
- Yasuhiro Manabe
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
299
|
Kim J, Alger BE. Inhibition of cyclooxygenase-2 potentiates retrograde endocannabinoid effects in hippocampus. Nat Neurosci 2004; 7:697-8. [PMID: 15184902 DOI: 10.1038/nn1262] [Citation(s) in RCA: 173] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2004] [Accepted: 04/06/2004] [Indexed: 11/08/2022]
Abstract
In hippocampal pyramidal cells, a rise in Ca(2+) releases endocannabinoids that activate the presynaptic cannabinoid receptor (CB1R) and transiently reduce GABAergic transmission-a process called depolarization-induced suppression of inhibition (DSI). The mechanism that limits the duration of endocannabinoid action in intact cells is unknown. Here we show that inhibition of cyclooxygenase-2 (COX-2), not fatty acid amide hydrolase (FAAH), prolongs DSI, suggesting that COX-2 limits endocannabinoid action.
Collapse
Affiliation(s)
- Jimok Kim
- Program in Neuroscience and Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | | |
Collapse
|
300
|
Gendron TF, Brunette E, Mealing GAR, Nguyen A, Tauskela JS, Morley P. Opposing effects of cyclooxygenase-2 selective inhibitors on oxygen-glucose deprivation-induced neurotoxicity. Eur J Pharmacol 2004; 493:45-55. [PMID: 15189763 DOI: 10.1016/j.ejphar.2004.04.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2004] [Accepted: 04/20/2004] [Indexed: 11/25/2022]
Abstract
Cyclooxygenase-2 inhibitors protect against excitotoxicity in vitro yet provide conflicting results in in vivo models of ischemia. To bridge the gap in understanding the discrepancies among these studies, the effects of different cyclooxygenase-2 inhibitors were studied in an in vitro model of ischemia. Oxygen-glucose deprivation (OGD) induced cyclooxygenase-2 protein expression in neuronal cortical cultures. Cyclooxygenase-2 inhibitors exhibited opposing effects on neuronal death induced by OGD. The acidic sulfonamides, N-(2-cyclohexyloxy-4-nitrophenyl) methanesulfonamide (NS-398) and N-(4-nitro-2-phenoxyphenyl)-methanesulfonamide (nimesulide), aggravated neuronal death by enhancing OGD-induced increases in extracellular glutamate and intracellular Ca2+ levels. In contrast, 1-[(4-methylsulfonyl)phenyl]-3-tri-fluoromethyl-5-(4-fluorophenyl)pyrazole (SC-58125) dose-dependently protected cultures against OGD by suppressing increases in extracellular glutamate and intracellular Ca2+ levels. The NS-398-induced aggravation of neuronal death was lost if the inhibitor was added only following the OGD. The timing of inhibitor application also determined its effects on N-methyl-D-aspartate (NMDA)-induced excitoxicity. NS-398 was protective when added both during and post-NMDA exposure, but not if NS-398 was also applied for 60 min prior to the insult. In contrast, SC-58125 afforded protection against NMDA in the presence or absence of a pre-incubation period. This study demonstrates that certain cyclooxygenase-2 inhibitors have opposing effects on neuronal survival depending on the timing of application and the nature of the insult. These results may account for the discrepancies among previous studies which used different inhibitors and different models of neurotoxicity.
Collapse
Affiliation(s)
- Tania F Gendron
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada K1H 8M5.
| | | | | | | | | | | |
Collapse
|