1
|
Iba M, Kim C, Kwon S, Szabo M, Horan-Portelance L, Peer CJ, Figg WD, Reed X, Ding J, Lee SJ, Rissman RA, Cookson MR, Overk C, Wrasidlo W, Masliah E. Inhibition of p38α MAPK restores neuronal p38γ MAPK and ameliorates synaptic degeneration in a mouse model of DLB/PD. Sci Transl Med 2023; 15:eabq6089. [PMID: 37163617 DOI: 10.1126/scitranslmed.abq6089] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/21/2023] [Indexed: 05/12/2023]
Abstract
Alterations in the p38 mitogen-activated protein kinases (MAPKs) play an important role in the pathogenesis of dementia with Lewy bodies (DLB) and Parkinson's disease (PD). Activation of the p38α MAPK isoform and mislocalization of the p38γ MAPK isoform are associated with neuroinflammation and synaptic degeneration in DLB and PD. Therefore, we hypothesized that p38α might be associated with neuronal p38γ distribution and synaptic dysfunction in these diseases. To test this hypothesis, we treated in vitro cellular and in vivo mouse models of DLB/PD with SKF-86002, a compound that attenuates inflammation by inhibiting p38α/β, and then investigated the effects of this compound on p38γ and neurodegenerative pathology. We found that inhibition of p38α reduced neuroinflammation and ameliorated synaptic, neurodegenerative, and motor behavioral deficits in transgenic mice overexpressing human α-synuclein. Moreover, treatment with SKF-86002 promoted the redistribution of p38γ to synapses and reduced the accumulation of α-synuclein in mice overexpressing human α-synuclein. Supporting the potential value of targeting p38 in DLB/PD, we found that SKF-86002 promoted the redistribution of p38γ in neurons differentiated from iPS cells derived from patients with familial PD (carrying the A53T α-synuclein mutation) and healthy controls. Treatment with SKF-86002 ameliorated α-synuclein-induced neurodegeneration in these neurons only when microglia were pretreated with this compound. However, direct treatment of neurons with SKF-86002 did not affect α-synuclein-induced neurotoxicity, suggesting that SKF-86002 treatment inhibits α-synuclein-induced neurotoxicity mediated by microglia. These findings provide a mechanistic connection between p38α and p38γ as well as a rationale for targeting this pathway in DLB/PD.
Collapse
Affiliation(s)
- Michiyo Iba
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Changyoun Kim
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Somin Kwon
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcell Szabo
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Liam Horan-Portelance
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cody J Peer
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - William D Figg
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xylena Reed
- Laboratory of Neurogenetics, Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jinhui Ding
- Laboratory of Neurogenetics, Computational Biology Group, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, and Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark R Cookson
- Laboratory of Neurogenetics, Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wolf Wrasidlo
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- Division of Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Panmand DS, Jishkariani D, Hall CD, Steel PJ, Asiri AM, Katritzky AR. Synthesis and Direct C2 Functionalization of Imidazolium and 1,2,4-Triazolium N-Imides. J Org Chem 2014; 79:10593-8. [DOI: 10.1021/jo501746u] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Deepak S. Panmand
- Center
for Heterocyclic Compounds, Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Davit Jishkariani
- Center
for Heterocyclic Compounds, Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200, United States
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - C. Dennis Hall
- Center
for Heterocyclic Compounds, Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Peter J. Steel
- Department
of Chemistry, University of Canterbury, Christchurch 8041, New Zealand
| | - Abdullah M. Asiri
- Department
of Chemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
- Center
of Excellence for Advanced Materials Research (CEAMR), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Alan R. Katritzky
- Center
for Heterocyclic Compounds, Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200, United States
- Department
of Chemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
| |
Collapse
|
3
|
Peripheral injection of SB203580 inhibits the inflammatory-dependent synthesis of proinflammatory cytokines in the hypothalamus. BIOMED RESEARCH INTERNATIONAL 2014; 2014:475152. [PMID: 24995301 PMCID: PMC4065737 DOI: 10.1155/2014/475152] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/09/2014] [Accepted: 05/20/2014] [Indexed: 11/20/2022]
Abstract
The study was designed to determine the effects of peripheral injection of SB203580 on the synthesis of interleukin- (IL-) 1β, IL-6, and tumor necrosis factor (TNF) α in the hypothalamus of ewes during prolonged inflammation. Inflammation was induced by the administration of lipopolysaccharide (LPS) (400 ng/kg) over 7 days. SB203580 is a selective ATP-competitive inhibitor of the p38 mitogen-activated protein kinase (MAPK), which is involved in the regulation of proinflammatory cytokines IL-1β, IL-6 and TNFα synthesis. Intravenous injection of SB203580 successfully inhibited (P < 0.01) synthesis of IL-1β and reduced (P < 0.01) the production of IL-6 in the hypothalamus. The p38 MAPK inhibitor decreased (P < 0.01) gene expression of TNFα but its effect was not observed at the level of TNFα protein synthesis. SB203580 also reduced (P < 0.01) LPS-stimulated IL-1 receptor type 1 gene expression. The conclusion that inhibition of p38 MAPK blocks LPS-induced proinflammatory cytokine synthesis seems to initiate new perspectives in the treatment of chronic inflammatory diseases also within the central nervous system. However, potential proinflammatory effects of SB203580 treatment suggest that all therapies using p38 MAPK inhibitors should be introduced very carefully with analysis of all expected and unexpected consequences of treatment.
Collapse
|
4
|
Denes A, Thornton P, Rothwell NJ, Allan SM. Inflammation and brain injury: acute cerebral ischaemia, peripheral and central inflammation. Brain Behav Immun 2010; 24:708-23. [PMID: 19770034 DOI: 10.1016/j.bbi.2009.09.010] [Citation(s) in RCA: 226] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 09/15/2009] [Accepted: 09/15/2009] [Indexed: 12/18/2022] Open
Abstract
Inflammation is a classical host defence response to infection and injury that has many beneficial effects. However, inappropriate (in time, place and magnitude) inflammation is increasingly implicated in diverse disease states, now including cancer, diabetes, obesity, atherosclerosis, heart disease and, most relevant here, CNS disease. A growing literature shows strong correlations between inflammatory status and the risk of cerebral ischaemia (CI, most commonly stroke), as well as with outcome from an ischaemic event. Intervention studies to demonstrate a causal link between inflammation and CI (or its consequences) are limited but are beginning to emerge, while experimental studies of CI have provided direct evidence that key inflammatory mediators (cytokines, chemokines and inflammatory cells) contribute directly to ischaemic brain injury. However, it remains to be determined what the relative importance of systemic (largely peripheral) versus CNS inflammation is in CI. Animal models in which CI is driven by a CNS intervention may not accurately reflect the clinical condition; stroke being typically induced by atherosclerosis or cardiac dysfunction, and hence current experimental paradigms may underestimate the contribution of peripheral inflammation. Experimental studies have already identified a number of potential anti-inflammatory therapeutic interventions that may limit ischaemic brain damage, some of which have been tested in early clinical trials with potentially promising results. However, a greater understanding of the contribution of inflammation to CI is still required, and this review highlights some of the key mechanism that may offer future therapeutic targets.
Collapse
Affiliation(s)
- A Denes
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
5
|
Munoz L, Ammit AJ. Targeting p38 MAPK pathway for the treatment of Alzheimer's disease. Neuropharmacology 2009; 58:561-8. [PMID: 19951717 DOI: 10.1016/j.neuropharm.2009.11.010] [Citation(s) in RCA: 294] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 10/30/2009] [Accepted: 11/24/2009] [Indexed: 10/20/2022]
Abstract
Accumulating evidence indicates that p38 mitogen-activated protein kinase (MAPK) could play more than one role in Alzheimer's disease (AD) pathophysiology and that patients suffering from AD dementia could benefit from p38 MAPK inhibitors. The p38 MAPK signalling has been widely accepted as a cascade contributing to neuroinflammation. However, deepening insight into the underlying biology of Alzheimer's disease reveals that p38 MAPK operates in other events related to AD, such as excitotoxicity, synaptic plasticity and tau phosphorylation. Although quantification of behavioural improvements upon p38 MAPK inhibition and in vivo evaluation of p38 MAPK significance to various aspects of AD pathology is still missing, the p38 MAPK is emerging as a new Alzheimer's disease treatment strategy. Thus, we present here an update on the role of p38 MAPK in neurodegeneration, with a focus on Alzheimer's disease, by summarizing recent literature and several key papers from earlier years.
Collapse
Affiliation(s)
- Lenka Munoz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The University of Sydney, NSW 2006, Australia.
| | | |
Collapse
|
6
|
Clarkson AN, Liu H, Schiborra F, Shaw O, Sammut IA, Jackson DM, Appleton I. Angiogenesis as a predictive marker of neurological outcome following hypoxia-ischemia. Brain Res 2007; 1171:111-21. [PMID: 17761153 DOI: 10.1016/j.brainres.2007.06.100] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 06/21/2007] [Accepted: 06/25/2007] [Indexed: 10/23/2022]
Abstract
Cerebral ischemia induces angiogenesis within and around infarcted tissue. The protection of existing and growth of new blood vessels may contribute to a more favorable outcome. The present study assessed whether angiogenesis can be used as a marker for neurodegeneration/neuroprotection in a model of hypoxia-ischemia (HI). Increased CD31 immunoreactivity 7 days post-HI indicated increased angiogenesis compared to controls (P<0.001). Treatment with the GABA(A) receptor modulator, clomethiazole (CMZ; 414 mg/kg/day), normalized the level of angiogenesis compared to HI + saline (P<0.001). Conversely, the non-selective nitric oxide synthase (NOS) inhibitor, L-NAME (5 mg/kg/day), markedly decreased angiogenesis compared to controls (P<0.001). Circulating plasma levels of IL-1alpha, IL-1beta and GM-CSF were significantly elevated post-HI. CMZ treatment attenuated these increases while also stimulating IL-10 levels. L-NAME treatment did not alter IL-1alpha or IL-1beta levels, but decreased endogenous IL-10 levels and exacerbated the ischemic lesion (P<0.001). CMZ treatment has been shown to increase NOS levels, while L-NAME halted the HI-induced increase in NOS activity (P<0.001). We conclude that angiogenesis can be used as a marker of neurodegeneration/neuroprotection for cerebral HI and is correlated to NOS activity and circulating inflammatory mediators.
Collapse
Affiliation(s)
- Andrew N Clarkson
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
7
|
Simi A, Lerouet D, Pinteaux E, Brough D. Mechanisms of regulation for interleukin-1beta in neurodegenerative disease. Neuropharmacology 2007; 52:1563-9. [PMID: 17428507 DOI: 10.1016/j.neuropharm.2007.02.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Revised: 02/27/2007] [Accepted: 02/28/2007] [Indexed: 11/27/2022]
Abstract
The interleukin-1 family of cytokines are central to the pathology of acute and chronic diseases of the central nervous system. We describe current evidence on the transcriptional and post-transcriptional regulation of interleukin-1beta production, secretion and activity in the brain. Regarding the induction of protein synthesis, the possible involvement of Toll like receptor-4 is discussed including evidence that ischemic brain damage is reduced in Toll like receptor-4 knockout mice. The post-translational involvement of the P2X7-receptor and caspase-1 in the processing and release of active IL-1beta is also considered, as is evidence suggesting a possible extracellular cleavage of pro-IL-1beta by neutrophil derived proteases. We provide some fresh perspectives on how interleukin-1beta may be regulated and how these mechanisms could be targeted in disease.
Collapse
Affiliation(s)
- Anastasia Simi
- Faculty of Life Sciences, C.2210 Michael Smith Building, University of Manchester, Manchester M13 9PT, UK
| | | | | | | |
Collapse
|
8
|
Clarkson AN. Anesthetic-mediated protection/preconditioning during cerebral ischemia. Life Sci 2007; 80:1157-75. [PMID: 17258776 DOI: 10.1016/j.lfs.2006.12.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 12/04/2006] [Accepted: 12/13/2006] [Indexed: 01/22/2023]
Abstract
Cerebral ischemia is a multi-faceted neurodegenerative pathology that causes cellular injury to neurons within the central nervous system. In light of the underlying mechanisms being elucidated, clinical trials to find possible neuroprotectants to date have failed, thus highlighting the need for new putative targets to offer protection. Recent evidence has clearly shown that anesthetics can confer significant protection and or induce a preconditioning effect against cerebral ischemia-induced injury. This review will focus on the putative protection/preconditioning that is afforded by anesthetics, their possible interaction with GABA(A) and glutamate receptors and two-pore potassium channels. In addition, the interaction with inflammatory, apoptotic and underlying molecular (particularly immediately early genes and inducible nitric oxide synthase etc) pathways, the activation of K(ATP) channels and the ability to provide lasting protection will also be addressed.
Collapse
Affiliation(s)
- Andrew N Clarkson
- Department of Anatomy and Structural Biology, University of Otago, PO Box 913, Dunedin 9054, New Zealand.
| |
Collapse
|
9
|
Clarkson AN, Clarkson J, Jackson DM, Sammut IA. Mitochondrial involvement in transhemispheric diaschisis following hypoxia-ischemia: Clomethiazole-mediated amelioration. Neuroscience 2006; 144:547-61. [PMID: 17112678 DOI: 10.1016/j.neuroscience.2006.09.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Revised: 09/22/2006] [Accepted: 09/22/2006] [Indexed: 11/24/2022]
Abstract
Mitochondria play a central role in both the physiological and pathophysiological regulation of cell survival/death. Increasing evidence places mitochondrial dysfunction at the center of many neuropathological conditions. The present study investigates the extent of mitochondrial dysfunction in cortical, hippocampal and cerebellar tissues in a rat model of hypoxia-ischemia (HI). We hypothesized that; mitochondrial dysfunction in situ may be prevented by treatment with clomethiazole (CMZ), a GABA(A) receptor agonist. Assessment of mitochondrial FAD-linked respiration at both 1- and 3-day post-HI revealed a marked decrease in activity from ipsilateral cortical and hippocampal regions (P<0.001). In addition, small changes were seen in contralateral cortical and hippocampal tissues as well as in the cerebellum at 3-days (P<0.05). Assessment of the mitochondrial electron transport chain (complexes I-V), and mitochondrial markers of integrity (citrate synthase) and oxidative stress (aconitase) confirmed mitochondrial impairment in ipsilateral regions following HI. Complexes I, II-III, V and citrate synthase were also impaired in contralateral regions and cerebellum 3-days post-HI. Treatment with CMZ (414 mg/kg/day via minipumps) provided marked protection to all aspects of neuronal tissue assessed. Circulating cytokine (interleukin [IL]-1alpha, IL-1beta, tumor necrosis factor [TNF]-alpha, granulocyte macrophage colony-stimulating factor [GM-CSF], IL-4 and IL-10) levels were also assessed in these animals 3-days post-HI. Plasma IL-1alpha, IL-1beta, TNF-alpha and GM-CSF levels were significantly increased post-HI. Treatment with CMZ ameliorated the increases in IL-1alpha, IL-1beta, TNF-alpha and GM-CSF levels while increasing plasma IL-4 and IL-10 levels. This study provides evidence of the extent of mitochondrial damage following an HI-insult. In addition, we have shown that protection afforded by CMZ extends to preservation of mitochondrial function and integrity via anti-inflammatory mediated pathways.
Collapse
Affiliation(s)
- A N Clarkson
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand.
| | | | | | | |
Collapse
|
10
|
Thornton P, Pinteaux E, Gibson RM, Allan SM, Rothwell NJ. Interleukin-1-induced neurotoxicity is mediated by glia and requires caspase activation and free radical release. J Neurochem 2006; 98:258-66. [PMID: 16805812 DOI: 10.1111/j.1471-4159.2006.03872.x] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Interleukin (IL)-1 expression is induced rapidly in response to diverse CNS insults and is a key mediator of experimentally induced neuronal injury. However, the mechanisms of IL-1-induced neurotoxicity are unknown. The aim of the present study was to examine the toxic effects of IL-1 on rat cortical cell cultures. Treatment with IL-1beta did not affect the viability of pure cortical neurones. However, IL-1 treatment of cocultures of neurones with glia or purified astrocytes induced caspase activation resulting in neuronal death. Neuronal cell death induced by IL-1 was prevented by pre-treatment with the IL-1 receptor antagonist, the broad spectrum caspase inhibitor Boc-Asp-(OMe)-CH(2)F or the antioxidant alpha-tocopherol. The NMDA receptor antagonist dizolcipine (MK-801) attenuated cell death induced by low doses of IL-1beta but the alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor antagonist 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo(F)quinoxaline (NBQX) had no effect. Inhibition of inducible nitric oxide synthase with N(omega)-nitro-l-arginine methyl ester had no effect on neuronal cell death induced by IL-1beta. Thus, IL-1 activates the IL-1 type 1 receptor in astrocytes to induce caspase-dependent neuronal death, which is dependent on the release of free radicals and may contribute to neuronal cell death in CNS diseases.
Collapse
Affiliation(s)
- Peter Thornton
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
11
|
Edling Y, Ingelman-Sundberg M, Simi A. Glutamate activatesc-fos in glial cells via a novel mechanism involving the glutamate receptor subtype mGlu5 and the transcriptional repressor DREAM. Glia 2006; 55:328-40. [PMID: 17120244 DOI: 10.1002/glia.20464] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Activation of c-fos in brain is related to coupling of neuronal activity to gene expression, but also to pathological conditions such as seizures or excitotoxicity-induced cell death. Glutamate activates c-fos in neurons through the calcium-dependent phosphorylation of CREB by ERK and/or CaMKIV kinase pathways downstream NMDA-receptors. In glial cells, however, the activation of c-fos by glutamate is poorly understood. Because glial cells actively modulate neuronal excitability and the brain's response to injury, we studied the mechanisms by which glutamate activates c-fos in rat cortical glial cells. Glutamate potently induced c-fos mRNA in a calcium-dependent manner, as demonstrated by using the calcium chelator BAPTA-AM. Glutamate-induced c-fos mRNA expression was not sensitive to inhibitors of ERK, p38(MAPK), or CaMK pathways, indicating that glial c-fos is activated by a distinct mechanism. Thapsigargin abolished the glutamate effect on c-fos mRNA, indicating ER calcium mobilization. Additionally, glutamate induction of c-fos mRNA was sensitive to the mGluR5 antagonist MPEP but not the NMDA-R antagonist MK-801. In luciferase reporter assays, DRE, which actively represses c-fos by binding the calcium-binding transcriptional repressor DREAM, was activated by glutamate, whereas SRE and CRE were not. Finally, glutamate caused the nuclear export of DREAM in astrocytes, and transfection of astrocytes with a mutant variant of DREAM that constitutively binds DNA inhibited glutamate-induced c-Fos expression. These findings are in sharp contrast to the mechanism described in neurons and suggest a novel pathway activated by glutamate in glial cells that employs mGluR5, ER calcium, and the derepression of c-fos at the DRE.
Collapse
Affiliation(s)
- Ylva Edling
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | | | | |
Collapse
|
12
|
Gatch MB, Selvig M, Forster MJ. GABAergic modulation of the discriminative stimulus effects of methamphetamine. Behav Pharmacol 2005; 16:261-6. [PMID: 15961966 PMCID: PMC3878065 DOI: 10.1097/01.fbp.0000166464.68186.25] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To assess whether gamma-aminobutyric acid (GABA) modulation of dopamine is important in mediation of the discriminative stimulus effects of methamphetamine, the GABA compounds chlordiazepoxide (benzodiazepine site agonist), pentobarbital (barbiturate site agonist), bicuculline and pentylenetetrazol (GABA(A) receptor antagonists) were tested in Sprague-Dawley rats trained to discriminate methamphetamine (1 mg/kg, i.p.) from saline. Each of the compounds produced modest amounts of methamphetamine-appropriate responding (20-35%) when tested alone. When tested in combination with methamphetamine, the antagonists (bicuculline and pentylenetetrazol) failed to shift the methamphetamine dose-effect curve. In contrast, chlordiazepoxide (25 mg/kg, i.p.) reduced methamphetamine-appropriate responding at each dose of methamphetamine tested, and pentobarbital (10 mg/kg, i.p.) dose-dependently decreased the discriminative stimulus effects of 1 mg/kg methamphetamine. In conclusion, GABA(A) antagonists and positive modulators likely do not produce methamphetamine-like stimulus effects. However, activation of GABA(A) receptors can interfere with the discriminative stimulus effects of methamphetamine.
Collapse
Affiliation(s)
- M B Gatch
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, 76107, USA.
| | | | | |
Collapse
|
13
|
Visser SAG, Pozarek S, Martinsson S, Forsberg T, Ross SB, Gabrielsson J. Rapid and long-lasting tolerance to clomethiazole-induced hypothermia in the rat. Eur J Pharmacol 2005; 512:139-51. [PMID: 15840398 DOI: 10.1016/j.ejphar.2005.02.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Revised: 02/04/2005] [Accepted: 02/22/2005] [Indexed: 11/19/2022]
Abstract
Mechanism, onset and duration of tolerance development to clomethiazole-induced hypothermia were investigated in rats using telemetry. The hypothermic effect of clomethiazole was completely abolished for 10 days after an s.c. injection of 300 micromol/kg and the effect returned to approximately 50% in 32 days. The gamma-aminobutyric acidA (GABA(A)) receptor agonist muscimol induced hypothermia at 88 micromol/kg without any (cross-) tolerance. GABA(A) receptor antagonists, bicuculline (5.4 micromol/kg) and picrotoxin (3.3 micromol/kg), did not inhibit clomethiazole-induced hypothermia nor the tolerance. The noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist, dizocilpine, counteracted clomethiazole-induced hypothermia at 3 micromol/kg but not the tolerance. Tolerance to the 5-hydroxytryptamine(1A) (5-HT(1A)) receptor agonist R-(+)-8-hydroxy-2-(di-n-propylamino)tetralin (R-8-OH-DPAT)-induced hypothermia was blocked by dizocilpine and clomethiazole but not vice versa. No pharmacokinetic interaction was observed. In conclusion, long-lasting tolerance to clomethiazole-induced hypothermia does not involve GABA(A) or 5-HT(1A) receptor functions. Glutamate via NMDA receptors may be involved in the hypothermic response but not in the tolerance.
Collapse
Affiliation(s)
- Sandra A G Visser
- PK/PD section, DMPK&BAC, Local Discovery Research Area CNS & Pain Control, AstraZeneca R&D Södertälje, SE-151 85 Södertälje, Sweden.
| | | | | | | | | | | |
Collapse
|
14
|
Clarkson AN, Liu H, Rahman R, Jackson DM, Appleton I, Kerr DS. Clomethiazole: mechanisms underlying lasting neuroprotection following hypoxia-ischemia. FASEB J 2005; 19:1036-8. [PMID: 15809357 DOI: 10.1096/fj.04-3367fje] [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: 11/11/2022]
Abstract
Damage after hypoxia-ischemia (HI) is observed in both cortical and subcortical regions. In this study, we employed a "Levine" rat model of HI (left carotid ligation + 1 h global hypoxia on PND-26) and used histological and electrophysiological paradigms to assess the long-term neuroprotective properties of clomethiazole (CMZ; a GABA(A) receptor modulator). Key enzymes involved in inflammation, namely nitric oxide synthase (NOS) and arginase, were also examined to assess potential CMZ mechanisms not involving GABA-R activation. Assessments were carried out 3 and 90 days post-HI. Extensive CNS lesions were evident after HI ipsilaterally at both short- and long-term intervals. CMZ significantly decreased the lesion size at 3 and 90 days (P<0.01; P<0.05). Evoked field potential analyses were used to assess hippocampal CA1 neuronal activity ex vivo. Electrophysiological measurements contralateral to the occlusion revealed impaired neuronal function after HI relative to short- and long-term controls (P<0.001, 3 and 14 days; P<0.01, 90 days), with CMZ treatment providing near complete protection (P<0.001 at 3 and 14 days; P<0.01 at 90 days). Both NOS and arginase activities were significantly increased at 3 days (P<0.01), with arginase remaining elevated at 90 days post-HI (P<0.05) ipsilaterally. CMZ suppressed the HI-induced increase in iNOS and arginase activities (P<0.001; P<0.05). These data provide evidence of long-term functional neuroprotection by CMZ in a model of HI. We further conclude that under conditions of HI, functional deficits are not restricted to the ipsilateral hemisphere and are due, at least in part, to changes in the activity of NOS and arginase.
Collapse
Affiliation(s)
- Andrew N Clarkson
- Department of Pharmacology and Toxicology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | | | | | | | | | | |
Collapse
|
15
|
Simi A, Edling Y, Ingelman-Sundberg M, Tindberg N. Activation of c-fos by lipopolysaccharide in glial cells via p38 mitogen-activated protein kinase-dependent activation of serum or cyclic AMP/calcium response element. J Neurochem 2005; 92:915-24. [PMID: 15686494 DOI: 10.1111/j.1471-4159.2004.02938.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pathological conditions such as ischaemic stroke and inflammatory disorders cause c-fos activation in the brain. This activation contributes to the initiation of the brain's inflammatory response, orchestrated by activated glial cells. The inflammatory signalling cascades leading to c-fos activation in glial cells are not well characterized. Thus, we have attempted a detailed analysis of the cis-acting elements, transcription factors and upstream kinase pathways involved in the activation of c-fos by lipopolysaccharide (LPS) in primary rat cortical glial cells. We found that (1) LPS-induced c-fos mRNA levels were sensitive to p38 mitogen-activated protein kinase (MAPK) inhibitors but not to mitogen-activated/extracellular signal-regulated kinase (ERK) or calcium-calmodulin-dependent kinase inhibitors, (2) LPS activated both serum response element (SRE) and cyclic AMP/calcium response element (CRE)-driven luciferase reporters in transient transfection assays, (3) LPS induced the phosphorylation of Elk1 CRE-binding protein (CREB)/activated transcription factor-1 (ATF-1) and the activation of GAL4-Elk1 and GAL4-CREB chimeric proteins, and (4) mutation of both SRE and CRE elements was necessary and sufficient to completely abolish LPS induction of a rat c-fos proximal promoter-luciferase reporter. Thus, c-fos activation by LPS in glial cells occurs via the SRE or CRE in an independent manner, and involves the Elk1 or CREB/ATF-1 transcription factors. Elk1-mediated transactivation was dependent on p38 MAPK, suggesting a crucial role of these factors in mediating inflammatory responses in the CNS.
Collapse
Affiliation(s)
- Anastasia Simi
- Institute of Environmental Medicine, Division of Molecular Toxicology, Karolinska Institute, Stockholm, Sweden.
| | | | | | | |
Collapse
|
16
|
Spangelo BL, Horrell S, Goodwin AL, Shroff S, Jarvis WD. Somatostatin and gamma-aminobutyric acid inhibit interleukin-1 beta-stimulated release of interleukin-6 from rat C6 glioma cells. Neuroimmunomodulation 2004; 11:332-40. [PMID: 15316244 DOI: 10.1159/000079414] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Accepted: 11/03/2003] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE We investigated the ability of inhibitory neurotransmitters to alter the interleukin-1 beta (IL-1 beta)-stimulated release of interleukin-6 (IL-6) from cultured glial tumor cells. METHODS C6 rat glioblastoma cells were exposed to either IL-1 beta or its putative second messenger lysophosphatidylcholine (LPC) in the absence or presence of the inhibitory neurotransmitters somatostatin (SRIF) or gamma-aminobutyric acid (GABA). Alternatively, C6 cells were pretreated with selective inhibitors of JNK or p38 and then exposed to either IL-1 beta or LPC to determine the relative involvement of these terminal stress kinases in the stimulation of IL-6 release. RESULTS IL-1 beta promoted the release of IL-6 with a maximally effective concentration of 25 ng/ml. Both SRIF-14 and SRIF-28 comparably suppressed stimulated IL-6 release with an ED(50) of approximately 50 nM. GABA also prevented IL-1 beta-driven IL-6 release (ED(50) = 100 microM). IL-1 beta and LPC synergistically enhanced release of IL-6 in the presence of the beta-adrenergic receptor agonist isoproterenol (ISO); these effects were largely reversed by SRIF or GABA. The pyridinylimidazole inhibitor of p38, SB-203580, completely blocked stimulation of IL-6 release by IL-1 beta or LPC; conversely, the anthrapyrazolone JNK inhibitor, SP-600125, was ineffective in modifying stimulated IL-6 release. CONCLUSIONS The effects of IL-1 beta and LPC on IL-6 release from glioma cells are effectively antagonized by the inhibitory neurotransmitters SRIF and GABA. On the basis of correlative studies, we propose that the ability of inhibitory transmitters such as SRIF and GABA to counter the induction of IL-6 release may entail suppression of p38 activity.
Collapse
Affiliation(s)
- Bryan L Spangelo
- Department of Chemistry, University of Nevada at Las Vegas, Las Vegas, NV 89154-4003, USA.
| | | | | | | | | |
Collapse
|
17
|
Kelicen P, Tindberg N. Lipopolysaccharide induces CYP2E1 in astrocytes through MAP kinase kinase-3 and C/EBPbeta and -delta. J Biol Chem 2003; 279:15734-42. [PMID: 14670949 DOI: 10.1074/jbc.m311850200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytochrome P450 2E1 (CYP2E1) is highly inducible in a subset of astrocytes in vivo following ischemic or mechanical injury and in vitro by lipopolysaccharide (LPS) or interleukin-1beta. We have studied the mechanism of induction, and found that transcriptional activation of CYP2E1 occurred within 3 h, and CYP2E1 dependent catalytic activity was induced more than 4-fold within 5 h. The induction was sensitive to several tyrosine kinase inhibitors, and was further modulated by inhibitors of p38 MAP kinase. MAP kinase kinase-3 (MKK3) was phosphorylated in response to LPS, and expression of constitutively active MKK3, but not the MAP kinase kinases MEKK1 or MKK1, activated CYP2E1. Transcriptional activation was mediated through a C/EBPbeta and -delta binding element situated at -486/-474, and appeared to involve activation of prebound factors as well as recruitment of newly synthesized C/EBPbeta and -delta. It is thus suggested that LPS induces MKK3 activation in astrocytes, which in turn stimulates a C/EBPbeta and -delta binding element to mediate transcriptional activation of CYP2E1.
Collapse
Affiliation(s)
- Pelin Kelicen
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institute, S-171-77 Stockholm, Sweden
| | | |
Collapse
|