51
|
Sweger EJ, Casper KB, Scearce-Levie K, Conklin BR, McCarthy KD. Development of hydrocephalus in mice expressing the G(i)-coupled GPCR Ro1 RASSL receptor in astrocytes. J Neurosci 2007; 27:2309-17. [PMID: 17329428 PMCID: PMC6673489 DOI: 10.1523/jneurosci.4565-06.2007] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We developed a transgenic mouse line that expresses the G(i)-coupled RASSL (receptor activated solely by synthetic ligand) Ro1 in astrocytes to study astrocyte-neuronal communication. Surprisingly, we found that all transgenics expressing Ro1 developed hydrocephalus. We analyzed these mice in an effort to develop a new model of hydrocephalus that will further our understanding of the pathophysiology of the disease. Expression of Ro1 was restricted to astrocytes by crossing the transgenic hGFAP-tTA (tet transactivator behind the human glial fibrillary acidic protein promoter) mouse line with the transgenic tetO-Ro1/tetO-LacZ mouse line. This cross produced double-transgenic mice that expressed Ro1 in astrocytes. All double transgenics developed hydrocephalus by postnatal day 15, whereas single-transgenic littermate controls appeared normal. Hydrocephalic Ro1 mice displayed enlarged ventricles, partial denudation of the ependymal cell layer, altered subcommissural organ morphology, and obliteration of the cerebral aqueduct. Severely hydrocephalic mice also had increased levels of phospho-Erk and GFAP expression. Administration of doxycycline to breeding pairs suppressed Ro1 expression and the onset of hydrocephalus in double-transgenic offspring. Ro1 animals maintained on dox did not develop hydrocephalus; however, if taken off doxycycline at weaning, double-transgenic mice developed enlarged ventricles within 7 weeks, indicating that Ro1 expression also induces hydrocephalus in adults. This study discovered a new model of hydrocephalus in which the rate of pathogenesis can be controlled enabling the study of the pathogenesis of both juvenile and adult onset hydrocephalus.
Collapse
Affiliation(s)
- Elizabeth J. Sweger
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, and
| | - Kristen B. Casper
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, and
| | - Kimberly Scearce-Levie
- Gladstone Institute of Cardiovascular Disease, University of California, San Francisco, California 94158
| | - Bruce R. Conklin
- Gladstone Institute of Cardiovascular Disease, University of California, San Francisco, California 94158
| | - Ken D. McCarthy
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, and
| |
Collapse
|
52
|
Choi JS, Kim HY, Cha JH, Lee MY. Ischemic preconditioning-induced activation of ERK1/2 in the rat hippocampus. Neurosci Lett 2006; 409:187-91. [PMID: 17029782 DOI: 10.1016/j.neulet.2006.09.053] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 09/15/2006] [Accepted: 09/15/2006] [Indexed: 11/19/2022]
Abstract
We investigated the activation and cellular localization of the extracellular signal-regulated kinases ERK1/2 in a rat model of ischemic tolerance induction. Adult male Sprague-Dawley rats were subjected to 3 min of sublethal ischemic preconditioning. Activation of ERK1/2 showed the characteristic time- and cell-dependent patterns. Rapid and short-lasting activation of ERK after 3 min of cerebral ischemia was noted immediately in the dentate granule cells and mossy fibers of the hippocampus, and then occurred sequentially in CA3 and CA1 neurons and dentate hilar neurons at 10 min. Phosphorylation of ERK1/2 in hippocampal neurons returned to the basal level in an ordered manner. Basal level phosphorylation was attained first, at 30 min, by the CA1 neurons, and was then observed in CA3 and granule cells by 1 h and noted in some dentate hilar neurons at 12 h. By contrast, phosphorylation of ERK1/2 in mossy fibers and the CA1 dendritic field was sustained for at least 3 d. Transient activation of ERK1/2 was induced also in astrocytes of the dentate hilar region at 1 d post-stimulation. These data demonstrate that the short cerebral-ischemic preconditioning induced rapid and transient activation of ERK1/2 in tolerance-acquired CA1 neurons as well as in ischemia-resistant CA3 and dentate granule cells, and that the short preconditioning sustained activation in mossy fibers and neuropil areas, suggesting that ERK1/2 activation may be involved in the mechanism of ischemic tolerance in the rat hippocampus.
Collapse
Affiliation(s)
- Jeong-Sun Choi
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, Seoul 137-701, South Korea
| | | | | | | |
Collapse
|
53
|
Webster B, Hansen L, Adame A, Crews L, Torrance M, Thal L, Masliah E. Astroglial activation of extracellular-regulated kinase in early stages of Alzheimer disease. J Neuropathol Exp Neurol 2006; 65:142-51. [PMID: 16462205 DOI: 10.1097/01.jnen.0000199599.63204.6f] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Characterization of the earliest neuropathologic features of Alzheimer disease (AD) indicates that synaptic degeneration accompanied by tau hyperphosphorylation and amyloid deposition might be an important feature. The mechanisms involved are unclear; however, dysregulation of signaling cascades such as the extracellular signal-regulated kinase (ERK) pathway might play a role. In this context, the main objective of this study was to determine whether ERK hyperactivation occurs in early stages of AD. We compared the patterns of total and phosphorylated ERK (pERK) expression in the midfrontal cortex of patients clinically and neuropathologically characterized with early, intermediate, or advanced AD. Immunocytochemical and Western blot analysis showed that in early AD, there was extensive activation of ERK in astroglial cells in the white matter accompanied by intense astrogliosis. In contrast, in patients with more advanced AD, pERK immunoreactivity was associated with neuronal cell bodies and dystrophic neurites around plaques. Levels of astroglial pERK immunoreactivity in the white matter were strongly correlated with scores of cognitive performance (Blessed, Mini-Mental Status Examination, and Clinical Dementia Rating) and with the severity of AD neuropathology (Braak stage). These findings suggest that astroglial ERK activation may be an important early response to the onset of AD pathology. Identification of cell signaling events unique to early AD may provide therapeutic targets for the prevention or delay of dementia.
Collapse
Affiliation(s)
- Brian Webster
- Department of Neurosciences, University of California-San Diego, La Jolla, CA 92093-0624, USA
| | | | | | | | | | | | | |
Collapse
|
54
|
Sharif A, Canton B, Junier MP, Chneiweiss H. PEA-15 Modulates TNFα Intracellular Signaling in Astrocytes. Ann N Y Acad Sci 2006; 1010:43-50. [PMID: 15033692 DOI: 10.1196/annals.1299.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PEA-15 is a small protein (15 kDa) that was first identified as an abundant phosphoprotein in brain astrocytes and subsequently shown to be widely expressed in different tissues and highly conserved among mammals. It is composed of an N-terminal death effector domain (DED) and a C-terminal tail of irregular structure. PEA-15 is regulated by multiple calcium-dependent phosphorylation pathways. PEA-15 is ideally positioned to play a major role in signal integration. Accordingly, it has been demonstrated that PEA-15 diverts astrocytes from TNFalpha-triggered apoptosis and regulates the actions of the ERK MAP kinase cascade by binding to ERK and altering its subcellular localization. Expression of PEA-15 directs TNFalpha outcomes toward survival, whereas its absence allows the development of the cytokine-induced cell death.
Collapse
Affiliation(s)
- Ariane Sharif
- INSERM U114, Department de Neuropharmacologie, Collège de France, 75231 Paris Cedex 05, France
| | | | | | | |
Collapse
|
55
|
Ramesh G, Philipp MT. Pathogenesis of Lyme neuroborreliosis: mitogen-activated protein kinases Erk1, Erk2, and p38 in the response of astrocytes to Borrelia burgdorferi lipoproteins. Neurosci Lett 2005; 384:112-6. [PMID: 15893422 DOI: 10.1016/j.neulet.2005.04.069] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2005] [Revised: 04/08/2005] [Accepted: 04/20/2005] [Indexed: 02/06/2023]
Abstract
Lyme borreliosis, which is prevalent both in the US and in Europe, is an infectious disease that may cause local inflammation in numerous organs. We have hypothesized that, as with some neurodegenerative diseases, the pathogenesis of the neurocognitive deficiencies associated with Lyme neuroborreliosis of the central nervous system also has an inflammatory component. Dysregulated production of pro-inflammatory cytokines such as IL-6 and TNF-alpha can lead to neuronal damage. Mitogen-activated protein kinases (MAPK) play a key role in the regulation of neuronal development, growth, and survival, as well as that of pro-inflammatory cytokine production. As a model, we explored the possibility that MAPK-mediated lipoprotein-induced apoptosis and gliosis of rhesus monkey astrocytes stimulated in vitro. Lipoproteins are the key inflammatory molecule type of Borrelia burgdorferi, the spirochete that causes Lyme disease, and we had previously shown that lipoprotein-induced TNF-alpha production in astrocytes caused astrocyte apoptosis, and IL-6 enhanced proliferation of these cells. Lipoproteins readily activated p38 and Erk1/2 MAPK, thus enlisting these pathways among the kinase pathways that spirochetes may address as they invade the central nervous system. We also investigated whether specific inhibition of p38 and Erk1/2 MAPK would inhibit TNF-alpha and IL-6 production and thus astrocyte apoptosis, and proliferation, respectively. Lipoprotein-stimulated IL-6 production was unaffected by the MAPK inhibitors. In contrast, inhibition of both p38 and Erk1/2 significantly diminished TNF-alpha production, and totally abrogated production of this cytokine when both MAPK pathways were inhibited simultaneously. MAPK inhibition thus may be considered as a strategy to control inflammation and apoptosis in Lyme neuroborreliosis.
Collapse
Affiliation(s)
- Geeta Ramesh
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, 18703 Three Rivers Road, Covington, LA 70433, USA
| | | |
Collapse
|
56
|
Hashimoto K, Parker A, Malone P, Gabelt BT, Rasmussen C, Kaufman PS, Hernandez MR. Long-term activation of c-Fos and c-Jun in optic nerve head astrocytes in experimental ocular hypertension in monkeys and after exposure to elevated pressure in vitro. Brain Res 2005; 1054:103-15. [PMID: 16081055 DOI: 10.1016/j.brainres.2005.06.050] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2005] [Revised: 06/10/2005] [Accepted: 06/14/2005] [Indexed: 11/30/2022]
Abstract
This study investigates whether the immediate early gene (IEG) products c-Fos and c-Jun are activated in vivo in monkeys with experimental glaucoma, and in vitro in cultured human ONH astrocytes exposed to hydrostatic pressure (HP). Three Rhesus monkeys with mild glaucomatous damage (mean intraocular pressure (IOP) 27 +/- 1.3 mm Hg approximately 42 weeks) and three with moderate glaucomatous damage (mean IOP 44 +/- 6.7% mm Hg approximately 11 weeks) were used for this study; the contralateral eye served as normal control (mean IOP 18.6 +/- 1.7 mm Hg). ONH tissues were stained with GFAP, DAPI, and c-Jun or c-Fos, and transcription factor positive and negative nuclei were counted to determine nuclear localization. Cultured human normal and glaucomatous ONH astrocytes exposed to elevated HP served as the in vitro model of elevated pressure. Activation and nuclear localization of c-Fos and c-Jun increased significantly in the monkeys with elevated IOP. These data correlated with axonal loss, reactive astrocytes, and remodeling of the optic disc. Cultured human ONH astrocytes showed increased nuclear localization of c-Fos and c-Jun under exposure to HP. Immunohistochemistry demonstrated that the upstream regulators of c-Fos and c-Jun, ERK-MAPK and MAPKp38 localized to the nuclei of ONH astrocytes in monkeys with experimental glaucoma. Taken together, these results demonstrate c-Fos and c-Jun activation in ONH astrocytes in vivo and in vitro, and that activation of both transcription factors is associated with ERK and MAPKp38 activation in experimental glaucoma, suggesting that activation of transcription factors may participate in the induction and maintenance of the reactive astrocyte phenotype in glaucomatous optic neuropathy.
Collapse
Affiliation(s)
- K Hashimoto
- Division of Ophthalmology and Visual Science, Niigata University Graduate School, 1-757 Asahimachi, Niigata 951-8510, Japan
| | | | | | | | | | | | | |
Collapse
|
57
|
Heffron DS, Mandell JW. Opposing roles of ERK and p38 MAP kinases in FGF2-induced astroglial process extension. Mol Cell Neurosci 2005; 28:779-90. [PMID: 15797724 DOI: 10.1016/j.mcn.2004.12.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2004] [Revised: 11/23/2004] [Accepted: 12/21/2004] [Indexed: 11/28/2022] Open
Abstract
The stellate processes of astroglial cells undergo extensive remodeling in response to neural injury. Little is known about intracellular signaling mechanisms controlling process extension. We tested roles for the ERK and p38 MAP kinase pathways in a simplified culture model. FGF2-induced process extension was preceded by a strong and transient phosphorylation of ERK, and a modest activation of p38 MAP kinase, which exhibited significant basal activity. Phosphorylated ERK was found predominantly in the cytoplasm, whereas activated p38 MAP kinase was nuclear. Process extension was completely blocked by the specific MEK inhibitor U0126. Conversely, inhibition of the p38 MAP kinase pathway with SB202190 stimulated spontaneous process growth and greatly potentiated FGF2-induced process extension. The p38 inhibitor effect was reproduced with an adenovirus expressing dominant-negative p38 MAP kinase. Selective pharmacological blockade of MAP kinase pathways may enable modulation of the astroglial response to injury so as to promote neural regeneration.
Collapse
Affiliation(s)
- Daniel S Heffron
- Department of Pathology, University of Virginia Health System, PO Box 800904, Charlottesville, VA 22908, USA
| | | |
Collapse
|
58
|
Milenkovic I, Nedeljkovic N, Filipovic R, Pekovic S, Culic M, Rakic L, Stojiljkovic M. Pattern of Glial Fibrillary Acidic Protein Expression Following Kainate-Induced Cerebellar Lesion in Rats. Neurochem Res 2005; 30:207-13. [PMID: 15895824 DOI: 10.1007/s11064-004-2443-9] [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] [Indexed: 10/25/2022]
Abstract
In the present study glial fibrillary acidic protein (GFAP) expression was assessed following intravermian injection of kainic acid (KA) or physiological saline to adult rat cerebellum. After 2- to 30-day recovery period, free-floating sections cut with a microtome were obtained and were proccessed for immunocytochemistry against GFAP. Injection of both kainate and physiological saline elicited significant astrogliotic reaction, i.e. in the area around the lesion thick GFAP-positive Bergmann fibers with typical orientation appeared in the molecular and hypertrophied astrocytes abundantly appeared in the granular layer. However, following kainate intoxication lesion was not surrounded by typical demarcation glial scar during 30-day recovery period in contrast to the appearance of usual glial scar in the group injected with physiological saline, as early as 7-day postlesion. Preserved spatial organization of Bergmann fibers and the absence of typical demarcating glial scar after kainate-induced cerebellar lesion suggest distinct pattern of astrogliosis that presents an interesting model system to study the importance of glial scar in the recovery after ischemic brain insults.
Collapse
Affiliation(s)
- I Milenkovic
- Department of Neurobiology and Neurochemistry, Institute of Biological Research Sinisa Stankovic, Serbia and Montenegro
| | | | | | | | | | | | | |
Collapse
|
59
|
Pannu R, Singh AK, Singh I. A novel role of lactosylceramide in the regulation of tumor necrosis factor alpha-mediated proliferation of rat primary astrocytes. Implications for astrogliosis following neurotrauma. J Biol Chem 2005; 280:13742-51. [PMID: 15668227 DOI: 10.1074/jbc.m411959200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The present study describes the role of glycosphingolipids in neuroinflammatory disease and investigates tumor necrosis factor alpha (TNFalpha)-induced astrogliosis following spinal cord injury. Astrogliosis is the hallmark of neuroinflammation and is characterized by proliferation of astrocytes and increased glial fibrillary acidic protein (GFAP) gene expression. In primary astrocytes, TNFalpha stimulation increased the intracellular levels of lactosylceramide (LacCer) and induced GFAP expression and astrocyte proliferation. D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol.HCl (PDMP), a glucosylceramide synthase and LacCer synthase (GalT-2) inhibitor, inhibited astrocyte proliferation and GFAP expression, which were reversed by exogenous supplementation of LacCer but not by other glycosphingolipids. TNFalpha caused a rapid increase in the activity of GalT-2 and synthesis of LacCer. Silencing of GalT-2 gene using antisense oligonucleotides also attenuated the proliferation of astrocytes and GFAP expression. The PDMP and antisense-mediated inhibition of proliferation and GFAP expression was well correlated with decreased Ras/ERK1/2 pathway activation. Furthermore, TNFalpha-mediated astrocyte proliferation and GFAP expression was also inhibited by LY294002, a phosphatidylinositol 3-kinase inhibitor, which was reversed by exogenous LacCer. LY294002 also inhibited TNFalpha-induced GalT-2 activation and LacCer synthesis, suggesting a phosphatidylinositol 3-kinase-mediated regulation of GalT-2. In vivo, PDMP treatment attenuated chronic ERK1/2 activation and spinal cord injury (SCI)-induced astrocyte proliferation with improved functional recovery post-SCI. Therefore, the in vivo studies support the conclusions drawn from cell culture studies and provide evidence for the role of LacCer in TNFalpha-induced astrogliosis in a rat model of SCI. To our knowledge, this is the first report demonstrating the role of LacCer in the regulation of TNFalpha-induced proliferation and reactivity of primary astrocytes.
Collapse
Affiliation(s)
- Ravinder Pannu
- Department of Pediatrics, Department of Pathology, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina and Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | |
Collapse
|
60
|
Ciccarelli R, D'Alimonte I, Santavenere C, D'Auro M, Ballerini P, Nargi E, Buccella S, Nicosia S, Folco G, Caciagli F, Di Iorio P. Cysteinyl-leukotrienes are released from astrocytes and increase astrocyte proliferation and glial fibrillary acidic protein via cys-LT1 receptors and mitogen-activated protein kinase pathway. Eur J Neurosci 2004; 20:1514-24. [PMID: 15355318 DOI: 10.1111/j.1460-9568.2004.03613.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Cysteinyl-leukotrienes (cys-LTs), potent mediators in inflammatory diseases, are produced by nervous tissue, but their cellular source and role in the brain are not very well known. In this report we have demonstrated that rat cultured astrocytes express the enzymes (5'-lipoxygenase and LTC(4) synthase) required for cys-LT production, and release cys-LTs in resting condition and, to a greater extent, in response to calcium ionophore A23187, 1 h combined oxygen-glucose deprivation or 2-methyl-thioATP, a selective P2Y(1)/ATP receptor agonist. MK-886, a LT synthesis inhibitor, prevented basal and evoked cys-LT release. In addition, 2-methyl-thioATP-induced cys-LT release was abolished by suramin, a P2 receptor antagonist, or by inhibitors of ATP binding cassette proteins involved in cys-LT release. We also showed that astrocytes express cys-LT(1) and not cys-LT(2) receptors. The stimulation of these receptors by LTD(4) activated the mitogen-activated protein kinase (MAPK) pathway. This effect was: (i) insensitive to inhibitors of receptor-coupled Gi protein (pertussis toxin) or tyrosine kinase receptors (genistein); (ii) abolished by MK-571, a cys-LT(1) selective receptor antagonist, or PD98059, a MAPK inhibitor; (iii) reduced by inhibitors of calcium/calmodulin-dependent kinase II (KN-93), Ca(2+)-dependent and -independent (GF102903X) or Ca(2+)-dependent (Gö6976) protein kinase C isoforms. LTD(4) also increased astrocyte proliferation and glial fibrillary acidic protein content, which are considered hallmarks of reactive astrogliosis. Both effects were counteracted by cell pretreatment with MK-571 or PD98059. Thus, cys-LTs released from astrocytes might play an autocrine role in the induction of reactive astrogliosis that, in brain injuries, contributes to the formation of a reparative glial scar.
Collapse
Affiliation(s)
- Renata Ciccarelli
- Department of Biomedical Sciences, Section of Pharmacology, Medical School, G. D'Annunzio University of Chieti, Chieti, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Barber SA, Uhrlaub JL, DeWitt JB, Tarwater PM, Zink MC. Dysregulation of mitogen-activated protein kinase signaling pathways in simian immunodeficiency virus encephalitis. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:355-62. [PMID: 14742241 PMCID: PMC1602250 DOI: 10.1016/s0002-9440(10)63125-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/10/2003] [Indexed: 10/18/2022]
Abstract
Central nervous system (CNS) disease is a frequent complication of human immunodeficiency virus (HIV)-1 infection. Identification of cellular mechanisms that control virus replication and that mediate development of HIV-associated neuropathology will provide novel strategies for therapeutic intervention. The milieu of the CNS during HIV infection is extraordinarily complex because of infiltration of inflammatory cells and production of chemokines, cytokines, and neurotoxic molecules. Cells in the CNS must integrate signaling pathways activated simultaneously by products of virus replication and infiltrating immune cells. In this study, we examined activation of mitogen-activated protein kinases (MAPKs) in the CNS of simian immunodeficiency virus-infected macaques during acute, asymptomatic, and terminal infection. We demonstrate that significantly increased (P < 0.02) activation of ERK MAPK, typically associated with anti-apoptotic and neuroprotective pathways, occurs predominantly in astrocytes and immediately precedes suppression of virus replication and macrophage activation that occur after acute infection. In contrast, significantly increased activation of proapoptotic, neurodegenerative MAPKs JNK (P = 0.03; predominantly in macrophages/microglia), and p38 (P = 0.03; predominantly in neurons and astrocytes) after acute infection correlates with subsequent resurgent virus replication and development of neurological lesions. This shift from classically neuroprotective to neurodegenerative MAPK pathways suggests that agents that inhibit activation of JNK/p38 may be protective against HIV-associated CNS disease.
Collapse
Affiliation(s)
- Sheila A Barber
- Department of Comparative Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA.
| | | | | | | | | |
Collapse
|
62
|
Mandell JW. Phosphorylation state-specific antibodies: applications in investigative and diagnostic pathology. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:1687-98. [PMID: 14578166 PMCID: PMC1892416 DOI: 10.1016/s0002-9440(10)63525-0] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Until recently, the investigation of protein phosphorylation was limited to biochemical studies of enzyme activities in homogenized tissues. The availability of hundreds of phosphorylation state-specific antibodies (PSSAs) now makes possible the study of protein phosphorylation in situ, and is opening many exciting opportunities in investigative and diagnostic pathology. This review illustrates the power of PSSAs, especially in immunohistochemical applications to human disease and animal models. Technical considerations, including antibody specificity and lability of phosphoepitopes, are covered, along with potential pitfalls, illustrated by a case study. In the arena of oncology, PSSAs may prove especially valuable in directly demonstrating the efficacy of chemotherapies targeted at protein kinase cascades. Novel applications of PSSAs are also beginning to reveal molecular mechanisms of inflammatory, degenerative, and toxin-induced diseases.
Collapse
Affiliation(s)
- James W Mandell
- Department of Pathology (Neuropathology) and Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
63
|
Choi JS, Kim SY, Park HJ, Cha JH, Choi YS, Kang JE, Chung JW, Chun MH, Lee MY. Upregulation of gp130 and differential activation of STAT and p42/44 MAPK in the rat hippocampus following kainic acid-induced seizures. ACTA ACUST UNITED AC 2003; 119:10-8. [PMID: 14597225 DOI: 10.1016/j.molbrainres.2003.08.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We investigated the activation and cellular distribution of two signaling pathways, the signal transducers and activators of transcription (STATs) and mitogen-activated protein kinases (MAPKs) following kainic acid (KA)-induced seizures, in relation to the expression of gp130, a common cytokine signal transducer for the interleukin (IL)-6 family of cytokines. Rapid and short-lasting upregulation of gp130 was observed in the granule cells. This became evident in astrocytes by 3 h, increased progressively to peak at 3 days, and was sustained for 10 days. STATs, including STAT1 and STAT3, and p42/44 MAPK were activated in distinct cellular and spatial distributions within the hippocampus following seizures. A rapid and sustained seizure-induced activation of STAT3 and STAT1, revealed by nuclear STAT3 and STAT1 immunoreactivities, was observed exclusively in reactive astrocytes in the hippocampus, nearly coinciding with the time course of gp130 expression; however, STAT3 activation was greater. In contrast, seizure induced the rapid and transient activation of p42/44 MAPK in a subpopulation of hippocampal neurons and in astrocytes, although with weaker staining intensity. Two signaling pathways involving gp130, STATs and MAPK, were differentially activated in reactive astrocytes after KA injection, indicating that STATs and MAPK may differentially mediate the astroglial reaction in the rat hippocampus after KA-induced seizures.
Collapse
Affiliation(s)
- Jeong-Sun Choi
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Shackelford DA, Yeh RY. Activation of extracellular signal-regulated kinases (ERK) during reperfusion of ischemic spinal cord. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 115:173-86. [PMID: 12877988 DOI: 10.1016/s0169-328x(03)00206-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The extracellular signal-regulated kinases (ERK) participate in numerous signaling pathways and are abundantly expressed in the CNS. It has been proposed that ERK activation promotes survival in models of neuronal injury. Inhibition of MEK, the upstream kinase that activates ERK, however, leads to neuroprotection in models of cerebral ischemia and trauma, suggesting that in this context ERK activation contributes to cellular damage. The effect of ischemia and reperfusion on activity and expression of ERK was investigated using a reversible model of rabbit spinal cord ischemia. Active ERK was observed in nai;ve animals, which decreased during 15 to 60 min of ischemia. Upon reperfusion, a robust activation of ERK was observed in animals occluded for 60 min that remained permanently paraplegic. Immunohistochemical analyses revealed increased staining of phosphorylated ERK (pERK) in glial cells and faint nuclear staining in motor neurons of animals occluded for 60 min and reperfused for 18 h. In contrast ERK activity did not increase in animals occluded for 15 min that regained motor function. No evidence of increased pERK immunoreactivity in motor neurons or nuclear translocation was noted in these animals. ERK1 was demonstrated to be identical to a p46 c-Jun/ATF-2 kinase previously shown to be activated by reperfusion after a 60-min occlusion. The results suggest that activation of ERK during reperfusion of ischemic spinal cord participates in the cellular pathways leading to neuronal damage.
Collapse
Affiliation(s)
- Deborah A Shackelford
- Department of Neurosciences, University of California at San Diego, La Jolla, CA 92093-0624, USA.
| | | |
Collapse
|
65
|
Shin T, Ahn M, Jung K, Heo S, Kim D, Jee Y, Lim YK, Yeo EJ. Activation of mitogen-activated protein kinases in experimental autoimmune encephalomyelitis. J Neuroimmunol 2003; 140:118-25. [PMID: 12864979 DOI: 10.1016/s0165-5728(03)00174-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The expression of mitogen-activated protein (MAP) kinases, including extracellular signal-regulated kinase (ERK), c-Jun NH(2)-terminal protein kinase (JNK), and p38, was analyzed in experimental autoimmune encephalomyelitis (EAE) in rats. Western blot analysis showed that the three MAP kinases (phosphorylated ERK (p-ERK), p-JNK, and p-p38) were increased significantly in the spinal cords of rats with EAE at the peak stage as compared with the levels in controls (p<0.05), and both p-ERK and p-JNK declined slightly in the recovery stage of EAE. Immunohistochemistry showed that p-ERK was constitutively expressed in brain cells, including astroglial cells, and showed enhanced immunoreactivity in those cells in EAE, while some T cells and macrophages were weakly immunopositive for p-ERK in EAE lesions. Both p-JNK and p-p38 were intensely immunostained in T cells in EAE lesions, while a few glial cells and astrocytes were weakly positive for both. Taking all these facts into consideration, we postulate that increased expression of the phosphorylated form of each MAP kinase plays an important role in the initiation of acute monophasic EAE. Differential expression of three MAP kinases was discerned in an animal model of human autoimmune central nervous system diseases, including multiple sclerosis.
Collapse
Affiliation(s)
- Taekyun Shin
- Department of Veterinary Medicine, Institute for Life Science, Cheju National University, 690-756, Cheju, South Korea.
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Carbonell WS, Mandell JW. Transient neuronal but persistent astroglial activation of ERK/MAP kinase after focal brain injury in mice. J Neurotrauma 2003; 20:327-36. [PMID: 12866812 DOI: 10.1089/089771503765172282] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Astrogliosis is a nearly ubiquitous response to a variety of insults to the central nervous system (CNS). This reaction is triggered rapidly, but can persist for years after the initial trauma. Little is known about the signaling mechanisms responsible for this activation and its chronic maintenance. Extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) activation is implicated in several functions important to the reactive glial phenotype such as cellular proliferation and motility. Here we utilize immunohistochemistry with a phosphorylation state-specific antibody (pERK) to characterize the temporal and spatial pattern of ERK/MAPK activation in neurons and glia following a forebrain stab lesion (FSL) in mice. Early activation (1 h) was primarily in perilesional neuronal elements, particularly of the hippocampus. Occasional perilesional glia were also positive for pERK. Additionally, ependymal cells bilaterally stained prominently for pERK. These patterns of pERK immunoreactivity at 1 h were abolished by pretreatment with the selective MEK inhibitor, SL327. ERK/MAPK activation at later time points between 1 day (d) and 30 d was primarily restricted to perilesional astrocytes with maximum labeling at 3 d. However, pERK-positive astrocytes represented only a subset of total GFAP-positive cells and were found more proximal to the lesion suggesting specific functional activation of these cells. Finally, immunostaining for the phosphorylated form of cAMP response element-binding (CREB) protein, a downstream target of the ERK/MAPK cascade, was increased in perilesional glia 7 d after FSL. Sustained activation of the ERK/MAPK signaling pathway in perilesional reactive glia suggests a critical role for this cascade in astrogliosis.
Collapse
Affiliation(s)
- W Shawn Carbonell
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | |
Collapse
|
67
|
Lennmyr F, Karlsson S, Gerwins P, Ata KA, Terént A. Activation of mitogen-activated protein kinases in experimental cerebral ischemia. Acta Neurol Scand 2002; 106:333-40. [PMID: 12460137 DOI: 10.1034/j.1600-0404.2002.01313.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Mitogen-activated protein kinases (MAPK) regulate cell survival and differentiation. The aim of the present study is to investigate the activation pattern of different MAPKs [extracellular signal-regulated kinase (ERK), c-jun-N-terminal kinase (JNK) and p38] after cerebral ischemia. MATERIAL AND METHODS Rats were subjected to cerebral ischemia using a model for transient (2 h) and permanent middle cerebral artery occlusion (MCAO). The rats were allowed 6 h to 1 week of survival before immunohistochemical evaluation with phospho-specific antibodies, recognizing activated MAPKs. RESULTS ERK was activated in ipsilateral blood vessels, neurons and glia, but also in contralateral vessels. JNK activation was absent in neurons but appeared in arterial blood vessels and glia at the lesion side. Active p38 was observed in macrophages in maturing infarcts. CONCLUSIONS ERK and JNK may participate in the angiogenic response to cerebral ischemia. ERK, but not JNK, was activated in neurons, possibly indicating a pathophysiologic role. Active p38 might be involved in the inflammatory reaction.
Collapse
Affiliation(s)
- F Lennmyr
- Department of Medical Sciences, Uppsala University Hospital, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
68
|
Otani N, Nawashiro H, Fukui S, Nomura N, Shima K. Temporal and spatial profile of phosphorylated mitogen-activated protein kinase pathways after lateral fluid percussion injury in the cortex of the rat brain. J Neurotrauma 2002; 19:1587-96. [PMID: 12542859 DOI: 10.1089/089771502762300247] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mitogen-activated protein kinases (MAPK) play a crucial role in signal transduction that regulates gene expression through transcriptional factor activity. The purpose of this study was to investigate the temporal expression and topographic distribution of the activated MAPK pathways including extracellular signal-regulated protein kinase (ERK), c-Jun NH(2)-terminal kinase (JNK), and p38 MAPK following traumatic brain injury (TBI) in the cortex of the rat brain. Adult male Sprague-Dawley rats (300-400 g) were subjected to lateral fluid percussion injury of moderate severity (3.5-4.0 atm) using the Dragonfly device model (no. HPD-1700). Phosphorylated-MAPK protein levels were quantified using Western blot analysis. Topographic distribution of immunoreactivity for phosphorylated-MAPK was examined using immunohistochemistry. Our findings showed that TBI significantly increased the phosphorylated-ERK (p-ERK) and -JNK (p-JNK) levels, but not the -p38 (p-p38) protein levels in the cortex surrounding the injury site. The immunoreactivity for p-ERK and p-JNK immediately after TBI were localized in neurons. The immunoreactivity for p-JNK was uniformly but only transiently induced and returned to control levels 1 h after TBI. The immunoreactivity for p-ERK was confirmed up until 30 min after TBI in the superficial neuronal layers. Double immunostaining using a glial-specific marker demonstrated that p-ERK was prominent in astrocytes 6 h after TBI. The current results suggest that the ERK and JNK pathways, but not the p38 MAPK pathways are involved in signal transduction in the cortex following TBI.
Collapse
Affiliation(s)
- Naoki Otani
- Department of Neurosurgery, National Defense Medical College, Tokorozawa, Japan.
| | | | | | | | | |
Collapse
|
69
|
Barber SA, Bruett L, Douglass BR, Herbst DS, Zink MC, Clements JE. Visna virus-induced activation of MAPK is required for virus replication and correlates with virus-induced neuropathology. J Virol 2002; 76:817-28. [PMID: 11752171 PMCID: PMC136850 DOI: 10.1128/jvi.76.2.817-828.2002] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is well accepted that viruses require access to specific intracellular environments in order to proliferate or, minimally, to secure future proliferative potential as latent reservoirs. Hence, identification of essential virus-cell interactions should both refine current models of virus replication and proffer alternative targets for therapeutic intervention. In the present study, we examined the activation states of mitogen-activated protein kinases (MAPKs), ERK-1/2, in primary cells susceptible to visna virus and report that virus infection induces and sustains activation of the ERK/MAPK pathway. Treatment of infected cells with PD98059, a specific inhibitor of the ERK/MAPK pathway, abolishes visna virus replication, as evidenced by extremely low levels of Gag protein expression and reverse transcriptase activity in culture supernatants. In addition, although visna virus-induced activation of MAPK is detectable within 15 min, early events of viral replication (i.e., reverse transcription, integration, and transcription) are largely unaffected by PD98059. Interestingly, further examination demonstrated that treatment with PD98059 results in decreased cytoplasmic expression of gag and env, but not rev, mRNA, highly suggestive of an ERK/MAPK-dependent defect in Rev function. In vivo analysis of ERK-1/2 activation in brains derived from visna virus-infected sheep demonstrates a strong correlation between ERK/MAPK activation and virus-associated encephalitis. Moreover, double-labeling experiments revealed that activation of MAPK occurs not only in cells classically infected by visna virus (i.e., macrophages and microglia), but also in astrocytes, cells not considered to be major targets of visna virus replication, suggesting that activation of the ERK/MAPK pathway may contribute to the virus-induced processes leading to neurodegenerative pathology.
Collapse
MESH Headings
- Animals
- Astrocytes/enzymology
- Astrocytes/virology
- Brain/enzymology
- Brain/pathology
- Brain/virology
- Butadienes/pharmacology
- Cells, Cultured
- Encephalitis, Viral/enzymology
- Encephalitis, Viral/pathology
- Encephalitis, Viral/virology
- Enzyme Activation/drug effects
- Enzyme Inhibitors/pharmacology
- Flavonoids/pharmacology
- Gene Products, env/biosynthesis
- Gene Products, env/genetics
- Gene Products, gag/biosynthesis
- Gene Products, gag/genetics
- Gene Products, rev/biosynthesis
- Gene Products, rev/genetics
- Goats/virology
- Immunohistochemistry
- MAP Kinase Kinase 1
- MAP Kinase Signaling System/drug effects
- Macrophages/enzymology
- Macrophages/virology
- Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Mitogen-Activated Protein Kinases/metabolism
- Nitriles/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Sheep/virology
- Virulence
- Virus Replication/drug effects
- Visna-maedi virus/drug effects
- Visna-maedi virus/genetics
- Visna-maedi virus/pathogenicity
- Visna-maedi virus/physiology
Collapse
Affiliation(s)
- Sheila A Barber
- Division of Comparative Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | |
Collapse
|
70
|
Zvalova D, Formstecher E, Fauquet M, Canton B, Chneiweiss H. Keeping TNF-induced apoptosis under control in astrocytes: PEA-15 as a 'double key' on caspase-dependent and MAP-kinase-dependent pathways. PROGRESS IN BRAIN RESEARCH 2001; 132:455-67. [PMID: 11545011 DOI: 10.1016/s0079-6123(01)32095-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- D Zvalova
- INSERM U114/Chaire de Neuropharmacologie, Collège de France, 75231 Paris, France
| | | | | | | | | |
Collapse
|
71
|
Zhu Y, Romero MI, Ghosh P, Ye Z, Charnay P, Rushing EJ, Marth JD, Parada LF. Ablation of NF1 function in neurons induces abnormal development of cerebral cortex and reactive gliosis in the brain. Genes Dev 2001; 15:859-76. [PMID: 11297510 PMCID: PMC312666 DOI: 10.1101/gad.862101] [Citation(s) in RCA: 455] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a prevalent genetic disorder that affects growth properties of neural-crest-derived cell populations. In addition, approximately one-half of NF1 patients exhibit learning disabilities. To characterize NF1 function both in vitro and in vivo, we circumvent the embryonic lethality of NF1 null mouse embryos by generating a conditional mutation in the NF1 gene using Cre/loxP technology. Introduction of a Synapsin I promoter driven Cre transgenic mouse strain into the conditional NF1 background has ablated NF1 function in most differentiated neuronal populations. These mice have abnormal development of the cerebral cortex, which suggests that NF1 has an indispensable role in this aspect of CNS development. Furthermore, although they are tumor free, these mice display extensive astrogliosis in the absence of conspicuous neurodegeneration or microgliosis. These results indicate that NF1-deficient neurons are capable of inducing reactive astrogliosis via a non-cell autonomous mechanism.
Collapse
MESH Headings
- Alleles
- Animals
- Cell Transformation, Neoplastic/genetics
- Cells, Cultured
- Cerebral Cortex/abnormalities
- Cerebral Cortex/embryology
- Disease Models, Animal
- Ganglia, Spinal/pathology
- Gene Expression Regulation, Developmental
- Genes, Neurofibromatosis 1
- Genes, Reporter
- Genes, Synthetic
- Genetic Vectors/genetics
- Gliosis/metabolism
- Integrases/genetics
- Integrases/physiology
- Lac Operon
- Learning Disabilities/genetics
- MAP Kinase Signaling System/genetics
- Mice
- Mice, Knockout
- Mice, Neurologic Mutants
- Mice, Transgenic
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- Neurofibromin 1
- Neurons/metabolism
- Promoter Regions, Genetic
- Proto-Oncogene Proteins p21(ras)/physiology
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/physiology
- Signal Transduction
- Synapsins/genetics
- Viral Proteins
- p120 GTPase Activating Protein/genetics
- p120 GTPase Activating Protein/physiology
Collapse
Affiliation(s)
- Y Zhu
- Center for Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9133, USA
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Mandell JW, Gocan NC, Vandenberg SR. Mechanical trauma induces rapid astroglial activation of ERK/MAP kinase: Evidence for a paracrine signal. Glia 2001; 34:283-95. [PMID: 11360301 DOI: 10.1002/glia.1062] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Astrogliosis is a prominent and ubiquitous reaction of astrocytes to many forms of CNS injury, often implicated in the poor regenerative capacity of the adult mammalian CNS. Transmembrane signals that rapidly trigger and maintain astroglial responses to injury are largely undefined. Several candidate inducers of astrogliosis, including growth factors and neuropeptides, act via the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway. We previously observed chronically activated ERK/MAPK in human reactive astrocytes. To investigate mechanisms of pathway activation in a defined in vitro model, primary cultured astroglial monolayers were subjected to focal mechanical injury. Within 2-10 min, ERK/MAPK was activated, but only in cells near the wound edge. By 30 min, the entire monolayer showed activation, which persisted for 4 to 8 h. ERK/MAPK activation was specifically blocked by application of the MEK inhibitors, PD98059 and U0126. Cell-cell contact was not necessary for intercellular spread of ERK/MAPK activation, and ERK/MAPK-stimulating activity was found in the injury-conditioned medium. The activating factor was shown to have a native size of 50-100 kD and did not signal through the classical EGF receptor. Injury-induced signaling to ERK/MAPK required Ras, as demonstrated by specific blockade after transient transfection with a dominant negative Ha-RasN17 construct. Finally, we demonstrated that focal lesioning of adult rat cortex induces a rapid activation and spreading of astroglial ERK/MAPK, suggesting that similar mechanisms may operate in astroglial activation following acute brain injury.
Collapse
Affiliation(s)
- J W Mandell
- Department of Pathology (Neuropathology), University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA.
| | | | | |
Collapse
|
73
|
Gass P, Kretz O, Wolfer DP, Berger S, Tronche F, Reichardt HM, Kellendonk C, Lipp HP, Schmid W, Schütz G. Genetic disruption of mineralocorticoid receptor leads to impaired neurogenesis and granule cell degeneration in the hippocampus of adult mice. EMBO Rep 2000; 1:447-51. [PMID: 11258486 PMCID: PMC1083761 DOI: 10.1093/embo-reports/kvd088] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To dissect the effects of corticosteroids mediated by the mineralocorticoid (MR) and the glucocorticoid receptor (GR) in the central nervous system, we compared MR-/- mice, whose salt loss syndrome was corrected by exogenous NaCI administration, with GR-/- mice having a brain-specific disruption of the GR gene generated by the Cre/loxP-recombination system. Neuropathological analyses revealed a decreased density of granule cells in the hippocampus of adult MR-/- mice but not in mice with disruption of GR. Furthermore, adult MR-/- mice exhibited a significant reduction of granule cell neurogenesis to 65% of control levels, possibly mediated by GR due to elevated corticosterone plasma levels. Neurogenesis was unaltered in adult mice with disruption of GR. Thus, we could attribute long-term trophic effects of adrenal steroids on dentate granule cells to MR. These MR-related alterations may participate in the pathogenesis of hippocampal changes observed in ageing, chronic stress and affective disorders.
Collapse
Affiliation(s)
- P Gass
- Division of Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Hebert MA, O'Callaghan JP. Protein phosphorylation cascades associated with methamphetamine-induced glial activation. Ann N Y Acad Sci 2000; 914:238-62. [PMID: 11085325 DOI: 10.1111/j.1749-6632.2000.tb05200.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Reactive gliosis is the most prominent response to diverse forms of central nervous system (CNS) injury. The signaling events that mediate this characteristic response to neural injury are under intense investigation. Several studies have demonstrated the activation of phosphoproteins within the mitogen-activated protein kinase (MAPK) and Janus kinase (JAK) pathways following neural insult. These signaling pathways may be involved or responsible for the glial response following injury, by virtue of their ability to phosphorylate and dynamically regulate the activity of various transcription factors. This study sought to delineate, in vivo, the relative contribution of MAPK- and JAK-signaling components to reactive gliosis as measured by induction of glial-fibrillary acidic protein (GFAP), following chemical-induced neural damage. At time points (6, 24, and 48 h) following methamphetamine (METH, 10 mg/kg x 4, s.c.) administration, female C57BL/6J mice were sacrificed by focused microwave irradiation, a technique that preserves steady-state phosphorylation. Striatal (target) and nontarget (hippocampus) homogenates were assayed for METH-induced changes in markers of dopamine (DA) neuron integrity as well as differences in the levels of activated phosphoproteins. GFAP upregulation occurred as early as 6 h, reaching a threefold induction 48 h following METH exposure. Neurotoxicant-induced reductions in striatal levels of DA and tyrosine hydroxylase (TH) paralleled the temporal profile of GFAP induction. Blots of striatal homogenates, probed with phosphorylation-state specific antibodies, demonstrated significant changes in activated forms of extracellular-regulated kinase 1/2 (ERK 1/2), c-jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK), MAPK/ERK kinase (MEK1/2), 70-kDa ribosomal S6 kinase (p70 S6), cAMP responsive element binding protein (CREB), and signal transducer and activator of transcription 3 (STAT3). MAPK-related phosphoproteins exhibited an activation profile that peaked at 6 h, remained significantly increased at 24, and fell to baseline levels 48 h following neurotoxicant treatment. The ribosomal S6 kinase was enhanced over 60% for all time points examined. Immunoreactivity profiles for the transcription factors CREB and STAT3 indicated maximal increases in phosphorylation occurring at 24 h, and measuring greater than 2- or 17-fold, respectively. Specific signaling events were found to occur with a time course suggestive of their involvement in the gliotic response. The toxicant-induced activation of these growth-associated signaling cascades suggests that these pathways could be obligatory for the triggering and/or persistence of reactive gliosis and may therefore serve as potential targets for modulation of glial response to neural damage.
Collapse
Affiliation(s)
- M A Hebert
- Department of Health & Human Services, Public Health Service, Centers for Disease Control and Prevention, Morgantown, West Virginia 26505-2888, USA
| | | |
Collapse
|
75
|
Slevin M, Krupinski J, Slowik A, Rubio F, Szczudlik A, Gaffney J. Activation of MAP kinase (ERK-1/ERK-2), tyrosine kinase and VEGF in the human brain following acute ischaemic stroke. Neuroreport 2000; 11:2759-64. [PMID: 10976958 DOI: 10.1097/00001756-200008210-00030] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We examined expression of vascular endothelial growth factor (VEGF), phosphorylation of mitogen activated protein kinase (MAP) kinase (ERK1 and ERK2) and tyrosine phosphorylation in 19 patients (aged 58-90 years; mean 75) who died 1-44 days after acute ischaemic stroke. In the grey matter penumbra, 13 of 19 patients showed an increase in MAP kinase tyrosine phosphorylation (ERK1; 2.0- to 8-fold, ERK2; 2.2- to 11-fold) compared with normal contralateral tissue. In almost all cases, ERK-2 phosphorylation was higher than ERK1. Of these 13 patients, 11 also showed a general increase in tyrosine kinase phosphorylation, and eight expressed increased levels of VEGF protein (2.5- to 5-fold). In tissue examined directly from the infarct core, activation of the above proteins was not observed in the, majority of patients. In the white matter, seven of 19 patients (penumbra), and nine of 19 patients (stroke) had an increase in MAP kinase tyrosine phosphorylation (ERK1; 2.0- to 4.6-fold and ERK-2; 2.3- to 5.4-fold respectively) compared with normal contralateral tissue. There was no relationship between activation of MAP kinase and expression of VEGF. Examination of phosphorylated MAP kinase by immunohistochemistry revealed an increase in immunoreactivity in neurones, astroglial cells, reactive microglia and endothelial cells in areas surrounding infarcts, especially in areas with the highest density of microvessels. In conclusion, chronic activation of tyrosine phosphorylated events, in particular redistribution and phosphorylation of MAP kinase (ERK1/ERK2) occurs consistently in the grey matter penumbra of brain tissue following ischaemic stroke, and may be associated with increase in expression of VEGF. These signal transduction events could be important determinants of the extent of neuronal survival and/or angiogenic activity in the recovering brain tissue.
Collapse
Affiliation(s)
- M Slevin
- Department of Biological Sciences, Manchester Metropolitan University, UK
| | | | | | | | | | | |
Collapse
|