201
|
The neuroprotective effects of the seeds of Cassia obtusifolia on transient cerebral global ischemia in mice. Food Chem Toxicol 2009; 47:1473-9. [DOI: 10.1016/j.fct.2009.03.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Revised: 03/11/2009] [Accepted: 03/24/2009] [Indexed: 11/20/2022]
|
202
|
Chaudhury S, Wadhwa S. Prenatal auditory stimulation alters the levels of CREB mRNA, p-CREB and BDNF expression in chick hippocampus. Int J Dev Neurosci 2009; 27:583-90. [PMID: 19559781 DOI: 10.1016/j.ijdevneu.2009.06.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2009] [Accepted: 06/17/2009] [Indexed: 01/23/2023] Open
Abstract
Prenatal auditory stimulation influences the development of the chick auditory pathway and the hippocampus showing an increase in various morphological parameters as well as expression of calcium-binding proteins. Calcium regulates the activity of cyclic adenosine monophosphate-response element binding (CREB) protein. CREB is known to play a role in development, undergo phosphorylation with neural activity as well as regulate transcription of BDNF. BDNF is important for the survival of neurons and regulates synaptic strength. Hence in the present study, we have evaluated the levels of CREB mRNA and protein along with p-CREB protein as well as BDNF mRNA and protein levels in the chick hippocampus at embryonic days (E) 12, E16, E20 and post-hatch day (PH) 1 following activation by prenatal auditory stimulation. Fertilized eggs were exposed to species-specific sound or sitar music (frequency range: 100-6300Hz) at 65dB levels for 15min/h over 24h from E10 till hatching. The control chick hippocampus showed higher CREB mRNA and p-CREB protein in the early embryonic stages, which later decline whereas BDNF mRNA and BDNF protein levels increase until PH1. The CREB mRNA and p-CREB protein were significantly increased at E12, E16 and PH1 in the auditory stimulated groups as compared to control group. A significant increase in the level of BDNF mRNA was observed from E12 and the protein expression from E16 onwards in both auditory stimulated groups. Therefore, enhanced phosphorylation of CREB during development following prenatal sound stimulation may be responsible for cell survival. Increased levels of p-CREB again at PH1 may trigger synthesis of proteins necessary for synaptic plasticity. Further, the increased levels of BDNF may also help in regulating synaptic plasticity.
Collapse
Affiliation(s)
- Sraboni Chaudhury
- Department of Anatomy, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | | |
Collapse
|
203
|
Giampà C, Middei S, Patassini S, Borreca A, Marullo F, Laurenti D, Bernardi G, Ammassari-Teule M, Fusco FR. Phosphodiesterase type IV inhibition prevents sequestration of CREB binding protein, protects striatal parvalbumin interneurons and rescues motor deficits in the R6/2 mouse model of Huntington's disease. Eur J Neurosci 2009; 29:902-10. [PMID: 19291221 DOI: 10.1111/j.1460-9568.2009.06649.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The phosphodiesterase type IV inhibitor rolipram increases cAMP response element-binding protein (CREB) phosphorylation and exerts neuroprotective effects in both the quinolinic acid rat model of Huntington's disease (DeMarch et al., 2007) and the R6/2 mouse including sparing of striatal neurons, prevention of neuronal intranuclear inclusion formation and attenuation of microglial reaction (DeMarch et al., 2008). In this study, we sought to determine if rolipram has a beneficial role in the altered distribution of CREB binding protein in striatal spiny neurons and in the motor impairments shown by R6/2 mutants. Moreover, we investigated whether rolipram treatment altered the degeneration of parvalbuminergic interneurons typical of Huntington's disease (Fusco et al., 1999). Transgenic mice and their wild-type controls from a stable colony maintained in our laboratory were treated with rolipram (1.5 mg/kg) or saline daily starting from 4 weeks of age. The cellular distribution of CREB binding protein in striatal spiny neurons was assessed by immunofluorescence, whereas parvalbuminergic neuron degeneration was evaluated by cell counts of immunohistochemically labeled tissue. Motor coordination and motor activity were also examined. We found that rolipram was effective in preventing CREB binding protein sequestration into striatal neuronal intranuclear inclusions, sparing parvalbuminergic interneurons of R6/2 mice, and rescuing their motor coordination and motor activity deficits. Our findings demonstrate the possibility of reversing pharmacologically the behavioral and neuropathological abnormalities of symptomatic R6/2 mice and underline the potential therapeutic value of phosphodiesterase type IV inhibitors in Huntington's disease.
Collapse
Affiliation(s)
- Carmela Giampà
- Laboratory of Neuroanatomy, Santa Lucia Foundation at the European Center for Brain Research, Via del Fosso Fiorano 64, 00143 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
204
|
McGuire J, Herman JP, Ghosal S, Eaton K, Sallee FR, Sah R. Acid-sensing by the T cell death-associated gene 8 (TDAG8) receptor cloned from rat brain. Biochem Biophys Res Commun 2009; 386:420-5. [PMID: 19501050 DOI: 10.1016/j.bbrc.2009.05.133] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Accepted: 05/31/2009] [Indexed: 10/20/2022]
Abstract
The T cell death-associated gene 8 (TDAG8) is a pH-sensing GPCR with a reported immune-specific expression profile. Here, we demonstrate pH-induced activation of TDAG8 receptor cloned from rodent brain (rTDAG8). Cloned rTDAG8 transcript showed 88-95% homology with human and mouse transcripts of lymphoid origin. RT-PCR revealed high expression of TDAG8 in forebrain limbic regions. Extracellular acidification induced significantly elevated intracellular cyclic AMP, and phosphorylated CREB in TDAG8 expressing cells. Acidification-induced LDH release was significantly attenuated in cells expressing TDAG8, suggesting neuroprotective potential against acidosis-related cell injury. Our results open up new areas of investigation into the relevance of TDAG8 in pH homeostasis and pathological states associated with acid-base dys-regulation in the brain such as ischemia and panic disorder.
Collapse
Affiliation(s)
- Jennifer McGuire
- Department of Psychiatry, University of Cincinnati, Genome Research Institute, 2170 East Galbraith Road, Cincinnati, OH 45237, USA
| | | | | | | | | | | |
Collapse
|
205
|
Discovery of transcriptional programs in cerebral ischemia by in silico promoter analysis. Brain Res 2009; 1272:3-13. [DOI: 10.1016/j.brainres.2009.03.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 03/09/2009] [Accepted: 03/19/2009] [Indexed: 12/19/2022]
|
206
|
Lin WY, Chang YC, Lee HT, Huang CC. CREB activation in the rapid, intermediate, and delayed ischemic preconditioning against hypoxic-ischemia in neonatal rat. J Neurochem 2009; 108:847-59. [PMID: 19183266 DOI: 10.1111/j.1471-4159.2008.05828.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ischemic preconditioning (IP) is a defense program in which exposure to sublethal ischemia followed by a period of reperfusion results in subsequent resistance to severe ischemic insults. Very few in vivo IP models have been established for neonatal brain. We examined whether rapid, intermediate, and delayed IP against hypoxic-ischemia (HI) could be induced in neonatal brain, and if so, whether the IP involved phosphorylation of cAMP response element-binding protein (pCREB) after HI. Postnatal day 7 rat pups were subjected to HI at 2 h (2-h IP), 6 h (6-h IP), or 22 h (22-h IP) after IP. We found all three IP groups had significantly reduced neuronal damage and TUNEL-(+) cells 24 h post-HI than no-IP group. Compared with control, the no-IP group had significant decreases of pCREB and mitochondria Bcl-2 levels in the ipsilateral cortex 24 h post-HI. In contrast, the three IP groups had increased pCREB and mitochondria Bcl-2 levels, and significant differences were found between three IP and no-IP groups. The increases of cleavage of caspase-3 and poly (ADP-ribose) polymerase and of cells with nuclear apoptosis inducing factor post-HI in no-IP group were all significantly reduced in three IP groups. The increases of caspase-3 and calpain-mediated proteolysis of a-spectrin post-HI were significantly reduced only in 22-h IP group. Furthermore, all three IP groups had long-term neuroprotection at behavioral and pathological levels compared with no-IP group. In conclusion, IP, rapid, intermediate, or delayed, in neonatal rat brain activates CREB, up-regulates Bcl-2, induces extensive brakes on caspase-dependent and -independent apoptosis after HI, and provides long-term neuroprotection.
Collapse
Affiliation(s)
- Wan-Ying Lin
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, Taiwan
| | | | | | | |
Collapse
|
207
|
Lee HT, Chang YC, Tu YF, Huang CC. VEGF-A/VEGFR-2 signaling leading to cAMP response element-binding protein phosphorylation is a shared pathway underlying the protective effect of preconditioning on neurons and endothelial cells. J Neurosci 2009; 29:4356-68. [PMID: 19357264 PMCID: PMC6665743 DOI: 10.1523/jneurosci.5497-08.2009] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 12/29/2008] [Accepted: 02/24/2009] [Indexed: 01/13/2023] Open
Abstract
Preconditioning protects endothelial cells as well as neurons from ischemic injury. In 7-d-old rat pups, ligating the carotid artery 1 h before hypoxia damaged the ipsilateral cerebral hemisphere; in contrast, ligating the artery 24 h before hypoxia provided complete neuroprotection. The protective effect of the 24 h artery ligation preconditioning model requires the activation of cAMP response element-binding protein (CREB). We tested the hypothesis that vascular endothelial growth factor (VEGF)-A/VEGF receptor-2 (VEGFR-2) signaling that leads to CREB activation is the shared pathway underlying the protective effect of preconditioning in neurons and endothelial cells. VEGF-A, VEGFR-1, or VEGFR-2 was inhibited by antisense oligodeoxynucleotides (ODNs) in vivo and by a VEGF-A neutralizing antibody or VEGFR-2 inhibitor in vitro. CREB phosphorylation (pCREB) and VEGF-A and VEGFR-2 expression were increased and colocalized in vascular endothelial cells and neurons in the ipsilateral cerebral cortex 24 h after ligation. The antisense ODN blockades of VEGF-A and VEGFR-2 decreased pCREB and reduced the protection of 24 h ligation preconditioning. Furthermore, oxygen-glucose deprivation (OGD) preconditioning upregulated VEGF-A, VEGFR-2, and pCREB levels and protected immortalized H19-7 neuronal cells and b.End3 vascular endothelial cells against 24 h OGD cell death. Blocking VEGF-A or VEGFR-2 reduced CREB activation and the effects of OGD preconditioning in neuronal cells and endothelial cells. Transfecting a serine-133 phosphorylation mutant CREB also inhibited the protective effect of OGD preconditioning. We conclude that VEGF-A/VEGFR-2 signaling leading to CREB phosphorylation is the shared pathway underlying the preconditioning-induced protective effect in neurons and vascular endothelial cells in the developing brain.
Collapse
Affiliation(s)
| | - Ying-Chao Chang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Yi-Fang Tu
- Institute of Clinical Medicine, and
- Department of Emergency Medicine, National Cheng Kung University Hospital, Tainan 70428, Taiwan, and
| | | |
Collapse
|
208
|
Yang S, Alkayed NJ, Hurn PD, Kirsch JR. Cyclic adenosine monophosphate response element-binding protein phosphorylation and neuroprotection by 4-phenyl-1-(4-phenylbutyl) piperidine (PPBP). Anesth Analg 2009; 108:964-70. [PMID: 19224810 PMCID: PMC2828492 DOI: 10.1213/ane.0b013e318192442c] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Previous studies show that the potent, prototypical sigma(1)-receptor agonist 4-phenyl-1-(4-phenylbutyl) piperidine (PPBP) prevents cell death after oxygen-glucose deprivation (OGD) in primary cortical neuronal cultures. We tested the hypothesis that PPBP protects neurons by a mechanism involving activation of the transcription factor cyclic adenosine monophosphate response element-binding protein (CREB). METHODS Primary cultured cortical neurons were exposed to 2 h of OGD and allowed to recover for 24 h, and PPBP treatment was initiated 15 min before the insult in the presence and absence of the sigma(1)-receptor antagonist rimcazole and inhibitors against protein kinases known to activate signal transduction cascades that result in CREB phosphorylation, such as H89 (protein kinase A inhibitor), LY294002 (PI3K inhibitor), U0126 (MEK1/2 inhibitor), or KN62 calmodulin kinase II inhibitor). Neuronal cell death was assayed by lactate dehydrogenase measurement 24 h after OGD. CREB phosphorylation was measured by immunoblot analysis at 30 min, 1 h, and 3 h of reoxygenation. Blots were quantitatively analyzed using Quantity One image analysis software. RESULTS PPBP increased CREB phosphorylation at 1 h after recovery from OGD, which was abolished by rimcazole (1.7 +/- 0.2 in PPBP and 0.8 +/- 0.1 in PPBP plus rimcazole with OGD compared with 0.9 +/- 0.1 in OGD alone, p-CREB/CREB). The PPBP-induced increase in CREB phosphorylation was blocked by H89 (0.5 +/- 0.07) but not U0126, KN62, or LY294002. PPBP treatment prevented OGD-induced cell death and pretreatment with H89 blocked this protection (0.18 +/- 0.02 in PPBP and 0.27 +/- 0.03 in PPBP plus H89 with OGD compared with 0.33 +/- 0.02 in OGD alone, lactate dehydrogenase assay). Pretreatment with LY294002, UO126, or KN62 had no effect on neuronal protection by PPBP. CONCLUSIONS These data suggest that the mechanism of neuroprotection by PPBP may be linked to CREB phosphorylation.
Collapse
Affiliation(s)
- Sufang Yang
- Department of Anesthesiology and Perioperative Medicine, Oregon Health Sciences University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239-3098, USA
| | | | | | | |
Collapse
|
209
|
Lee B, Cao R, Choi YS, Cho HY, Rhee AD, Hah CK, Hoyt KR, Obrietan K. The CREB/CRE transcriptional pathway: protection against oxidative stress-mediated neuronal cell death. J Neurochem 2009; 108:1251-65. [PMID: 19141071 DOI: 10.1111/j.1471-4159.2008.05864.x] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Formation of reactive oxygen and nitrogen species is a precipitating event in an array of neuropathological conditions. In response to excessive reactive oxygen species (ROS) levels, transcriptionally dependent mechanisms drive the up-regulation of ROS scavenging proteins which, in turn, limit the extent of brain damage. Here, we employed a transgenic approach in which cAMP-response element binding protein (CREB)-mediated transcription is repressed (via A-CREB) to examine the contribution of the CREB/cAMP response element pathway to neuroprotection and its potential role in limiting ROS toxicity. Using the pilocarpine-evoked repetitive seizure model, we detected a marked enhancement of cell death in A-CREB transgenic mice. Paralleling this, there was a dramatic increase in tyrosine nitration (a marker of reactive species formation) in A-CREB transgenic mice. In addition, inducible expression of peroxisome proliferator-activated receptor gamma coactivator-1alpha was diminished in A-CREB transgenic mice, as was activity of complex I of the mitochondrial electron transport chain. Finally, the neuroprotective effect of brain-derived neurotrophic factor (BDNF) against ROS-mediated cell death was abrogated by disruption of CREB-mediated transcription. Together, these data both extend our understanding of CREB functionality and provide in vivo validation for a model in which CREB functions as a pivotal upstream integrator of neuroprotective signaling against ROS-mediated cell death.
Collapse
Affiliation(s)
- Boyoung Lee
- Department of Neuroscience, Ohio State University, Columbus, 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
210
|
Ward RJ, Lallemand F, de Witte P. Biochemical and neurotransmitter changes implicated in alcohol-induced brain damage in chronic or 'binge drinking' alcohol abuse. Alcohol Alcohol 2009; 44:128-35. [PMID: 19155229 DOI: 10.1093/alcalc/agn100] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The brain damage, which occurs after either chronic alcoholization or binge drinking regimes, shows distinct biochemical and neurotransmitter differences. An excessive amount of glutamate is released into specific brain regions during binge drinking (in excess of 4- to 5-fold of the normal basal concentration) that is not evident during periods of excessive alcohol consumption in chronic alcohol abusers. Increases in glutamate release are only observed during the initial stages of withdrawal from chronic alcoholism ( approximately 2- to 3-fold) due to alterations in the sensitivities of the NMDA receptors. Such changes in either density or sensitivity of these receptors are reported to be unaltered by binge drinking. When such excesses of glutamate are released in these two different models of alcohol abuse, a wide range of biochemical changes occur, mediated in part by increased fluxes of calcium ions and/or activation of various G-protein-associated signalling pathways. Cellular studies of alveolar macrophages isolated from these two animal models of alcohol abuse showed enhanced (binge drinking) or reduced (chronic alcoholization) lipopolysaccharide (LPS)-stimulated NO release. Such studies could suggest that neuroadaptation occurs with the development of tolerance to alcohol's effects in both neurotransmitter function and cellular processes during chronic alcoholization that delay the occurrence of brain damage. In contrast, 'binge drinking' induces immediate and toxic effects and there is no evidence of an increased preference for alcohol as seen after withdrawal from chronic alcoholization.
Collapse
Affiliation(s)
- Roberta J Ward
- Biologie du Comportement, Université Catholique de Louvain, Louvain-la-Neuve, Belgium.
| | | | | |
Collapse
|
211
|
Pittenger C, Coric V, Banasr M, Bloch M, Krystal JH, Sanacora G. Riluzole in the treatment of mood and anxiety disorders. CNS Drugs 2008; 22:761-86. [PMID: 18698875 DOI: 10.2165/00023210-200822090-00004] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent advances implicate amino acid neurotransmission in the pathophysiology and treatment of mood and anxiety disorders. Riluzole, which is approved and marketed for the treatment of amyotrophic lateral sclerosis, is thought to be neuroprotective through its modulation of glutamatergic neurotransmission. Riluzole has multiple molecular actions in vitro; the two that have been documented to occur at physiologically realistic drug concentrations and are therefore most likely to be clinically relevant are inhibition of certain voltage-gated sodium channels, which can lead to reduced neurotransmitter release, and enhanced astrocytic uptake of extracellular glutamate.Although double-blind, placebo-controlled trials are lacking, several open-label trials have suggested that riluzole, either as monotherapy or as augmentation of standard therapy, reduces symptoms of obsessive-compulsive disorder, unipolar and bipolar depression, and generalized anxiety disorder. In studies of psychiatrically ill patients conducted to date, the drug has been quite well tolerated; common adverse effects include nausea and sedation. Elevation of liver function tests is common and necessitates periodic monitoring, but has been without clinical consequence in studies conducted to date in psychiatric populations. Case reports suggest utility in other conditions, including trichotillomania and self-injurious behaviour associated with borderline personality disorder. Riluzole may hold promise for the treatment of several psychiatric conditions, possibly through its ability to modulate pathologically dysregulated glutamate levels, and merits further investigation.
Collapse
Affiliation(s)
- Christopher Pittenger
- Clinical Neuroscience Research Unit, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | | | | | |
Collapse
|
212
|
Song MS, Rauw G, Baker GB, Kar S. Memantine protects rat cortical cultured neurons against β-amyloid-induced toxicity by attenuating tau phosphorylation. Eur J Neurosci 2008; 28:1989-2002. [DOI: 10.1111/j.1460-9568.2008.06498.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
213
|
Crews FT, Nixon K. Mechanisms of neurodegeneration and regeneration in alcoholism. Alcohol Alcohol 2008; 44:115-27. [PMID: 18940959 DOI: 10.1093/alcalc/agn079] [Citation(s) in RCA: 408] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIMS This is a review of preclinical studies covering alcohol-induced brain neuronal death and loss of neurogenesis as well as abstinence-induced brain cell genesis, e.g. brain regeneration. Efforts are made to relate preclinical studies to human studies. METHODS The studies described are preclinical rat experiments using a 4-day binge ethanol treatment known to induce physical dependence to ethanol. Neurodegeneration and cognitive deficits following binge treatment mimic the mild degeneration and cognitive deficits found in humans. Various histological methods are used to follow brain regional degeneration and regeneration. RESULTS Alcohol-induced degeneration occurs due to neuronal death during alcohol intoxication. Neuronal death is related to increases in oxidative stress in brain that coincide with the induction of proinflammatory cytokines and oxidative enzymes that insult brain. Degeneration is associated with increased NF-kappaB proinflammatory transcription and decreased CREB transcription. Corticolimbic brain regions are most sensitive to binge-induced degeneration and induce relearning deficits. Drugs that block oxidative stress and NF-kappaB transcription or increase CREB transcription block binge-induced neurodegeneration, inhibition of neurogenesis and proinflammatory enzyme induction. Regeneration of brain occurs during abstinence following binge ethanol treatment. Bursts of proliferating cells occur across multiple brain regions, with many new microglia across brain after months of abstinence and many new neurons in neurogenic hippocampal dentate gyrus. Brain regeneration may be important to sustain abstinence in humans. CONCLUSIONS Alcohol-induced neurodegeneration occurs primarily during intoxication and is related to increased oxidative stress and proinflammatory proteins that are neurotoxic. Abstinence after binge ethanol intoxication results in brain cell genesis that could contribute to the return of brain function and structure found in abstinent humans.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | |
Collapse
|
214
|
Lee JH, Park SY, Shin HK, Kim CD, Lee WS, Hong KW. Protective effects of cilostazol against transient focal cerebral ischemia and chronic cerebral hypoperfusion injury. CNS Neurosci Ther 2008; 14:143-52. [PMID: 18482026 DOI: 10.1111/j.1527-3458.2008.00042.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Cilostazol increases intracellular cyclic adenosine monophosphate (cyclic AMP) levels by inhibiting type III phosphodiesterase. It was approved by the Food and Drug Administration for the treatment of intermittent claudication. Its principal actions include inhibition of platelet aggregation, antithrombotic action in cerebral ischemia, and vasodilation, mediated by increased cyclic AMP levels. In a multicenter, randomized, placebo-controlled, double-blind clinical trial, cilostazol has been shown to protect patients from recurrent cerebral infarction. It has been recently suggested that cilastozol could be useful in the treatment of transient focal cerebral ischemic injury. Beneficial effects of cilostazol in cerebral ischemic infarction and edema formation has been confirmed in rats by the magnetic resonance imaging (MRI). The preventive effect was ascribed to cAMP-dependent protein kinase (PKA)-coupled maxi-K channel activation with additional antioxidant and poly(adenosine diphosphate [ADP]-ribose) polymerase inhibitory actions. Most recently, cilostazol has been shown to prevent vacuolation and rarefaction in the white matter of the rats subjected to chronic cerebral hypoperfusion in association with suppression of astrocyte and microglial activation. Taken together, recent experimental studies with cilostazol showed promising results in cerebral ischemia and chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Jeong Hyun Lee
- Department of Pharmacology, College of Medicine, Pusan National University, Busan, Korea
| | | | | | | | | | | |
Collapse
|
215
|
Abstract
A hallmark of glaucomatous optic nerve damage is retinal ganglion cell (RGC) death. RGCs, like other central nervous system neurons, have a limited capacity to survive or regenerate an axon after injury. Strategies that prevent or slow down RGC degeneration, in combination with intraocular pressure management, may be beneficial to preserve vision in glaucoma. Recent progress in neurobiological research has led to a better understanding of the molecular pathways that regulate the survival of injured RGCs. Here we discuss a variety of experimental strategies including intraocular delivery of neuroprotective molecules, viral-mediated gene transfer, cell implants and stem cell therapies, which share the ultimate goal of promoting RGC survival after optic nerve damage. The challenge now is to assess how this wealth of knowledge can be translated into viable therapies for the treatment of glaucoma and other optic neuropathies.
Collapse
|
216
|
Functional interference between glycogen synthase kinase-3 beta and the transcription factor Nrf2 in protection against kainate-induced hippocampal cell death. Mol Cell Neurosci 2008; 39:125-32. [PMID: 18619545 DOI: 10.1016/j.mcn.2008.06.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Revised: 05/19/2008] [Accepted: 06/10/2008] [Indexed: 12/18/2022] Open
Abstract
Excitotoxicity mediated by glutamate receptors may underlay the pathology of several neurologic diseases. Considering that oxidative stress is central to excitotoxic damage, in this study we sought to analyze if the transcription factor Nrf2, guardian of redox homeostasis, might be targeted to prevent kainate-induced neuron death. Hippocampal slices from Nrf2 knockout mice exhibited increased oxidative stress and cell death compared to those of control mice in response to kainate, as determined with the redox sensitive probes 2,7-dichlorodihydrofluorescein diacetate (H(2)DCFAC) and propidium iodide and lactate dehydrogenase release, respectively, therefore demonstrating a role of Nrf2 in antioxidant protection against excitotoxicity. In the hippocampus of mice intraperitoneally injected with kainate we observed a rapid activation of Akt, inhibition of GSK-3beta and translocation of Nrf2 to the nucleus, but after 4 h Akt was inactive, GSK-3beta was active and Nrf2 was mostly cytosolic, therefore extending our previous studies which indicate that GSK-3beta excludes Nrf2 from the nucleus. Lithium, a GSK-3beta inhibitor, promoted Nrf2 transcriptional activity towards an Antioxidant-Response-Element (ARE) luciferase reporter and cooperated with sulforaphane (SFN) to induce this reporter and to increase the protein levels of heme oxygenase-1 (HO-1), coded by a representative ARE-containing gene. Conversely, ARE activation by SFN was attenuated by over-expression of active GSK-3beta. Finally, combined treatment with SFN and lithium attenuated oxidative stress and cell death in kainate-treated hippocampal slices of wild type mice but not Nrf2 null littermates. Our findings identify the axis GSK-3beta/Nrf2 as a pharmacological target in prevention of excitotoxic neuronal death.
Collapse
|
217
|
Rybnikova E, Gluschenko T, Tulkova E, Churilova A, Jaroshevich O, Baranova K, Samoilov M. Preconditioning induces prolonged expression of transcription factors pCREB and NF-kappa B in the neocortex of rats before and following severe hypobaric hypoxia. J Neurochem 2008; 106:1450-8. [PMID: 18547368 DOI: 10.1111/j.1471-4159.2008.05516.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Preconditioning using mild repetitive hypobaric hypoxia is known to increase a tolerance of brain neurons to severe hypoxia and other injurious exposures. In the present study, the effects of mild hypoxic preconditioning on the expression of transcription factors NF-kappaB and phosphorylated CREB (pCREB) has been studied in the neocortex of rats exposed to severe hypobaric hypoxia. As revealed by quantitative immunocytochemistry, the injurious severe hypobaric hypoxia (180 Torr, 3 h) remarkably reduced the neocortical levels of pCREB and NF-kappaB. The three-trial hypoxic preconditioning (360 Torr, 2 h, 3 days) induced persistent up-regulation of pCREB and NF-kappaB expression in the neocortex of rats 3-24 h following the severe hypoxia. In addition, the preconditioning alone which was not followed by the severe hypoxia, considerably increased neocortical pCREB and NF-kappaB levels. The findings suggest a role for transcription factors cAMP response element-binding protein and NF-kappaB in the neuroprotective mechanisms activated by the hypoxic preconditioning.
Collapse
Affiliation(s)
- Elena Rybnikova
- Laboratory of Neuroendocrinology, Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | | | | | | | | | | | | |
Collapse
|
218
|
O'Neill C. Phosphatidylinositol 3-kinase signaling in mammalian preimplantation embryo development. Reproduction 2008; 136:147-56. [PMID: 18515313 DOI: 10.1530/rep-08-0105] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The development of the preimplantation mammalian embryo is an autopoietic process; once initiated development proceeds without an absolute requirement for external information or growth cues. This developmental autonomy is partly explained by the generation of autocrine trophic ligands that are released and act back on the embryo via specific receptors. Several embryotrophic ligands cause receptor-dependent activation of 1-o-phosphatidylinositol 3-kinase. This enzyme phosphorylates phosphatidylinositol-4,5-bisphosphate to form phosphatidylinositol-3,4,5-trisphosphate. Genetic or pharmacological ablation of this enzyme activity disrupts normal development of preimplantation embryos. Phosphatidylinositol-3,4,5-trisphosphate is a membrane lipid that acts as a docking site for a wide range of proteins possessing the pleckstrin homology (PH) domain. Such proteins are important regulators of cell survival, proliferation, and differentiation. RAC-alpha serine/threonine protein kinase is an important PH domain protein and its activity is required for normal preimplantation embryo development and survival. The activity of a range of PH domain proteins is also implicated in the normal development of the embryo. This review critically examines the evidence for the activation of 1-o-phosphatidylinositol 3-kinase in the generation of pleiotypic trophic response to embryotrophins in the autopoietic development of the preimplantation embryo.
Collapse
Affiliation(s)
- Chris O'Neill
- Disciplines of Medicine and Physiology, Human Reproduction Unit, Royal North Shore Hospital, University of Sydney, St Leonards, New South Wales 2065, Australia.
| |
Collapse
|
219
|
Beneficial effects of rolipram in the R6/2 mouse model of Huntington's disease. Neurobiol Dis 2008; 30:375-387. [PMID: 18424161 DOI: 10.1016/j.nbd.2008.02.010] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Revised: 02/08/2008] [Accepted: 02/22/2008] [Indexed: 12/24/2022] Open
Abstract
We have previously showed that rolipram, a phosphodiesterase type IV inhibitor, displays a neuroprotective effect in a rat quinolinic acid model of HD [DeMarch Z., Giampa C., Patassini S., Martorana A., Bernardi G. and Fusco F.R., (2007) Beneficial effects of rolipram in a quinolinic acid model of striatal excitotoxicity. Neurobiol. Dis. 25:266-273.]. In this study, we sought to determine if rolipram exerts a neuroprotective effect in R6/2 mutant mice, which recapitulates, in many aspects, human HD [Mangiarini L., Sathasivam K., Seller M., Cozens B., Harper A., Hetherington C., Lawton M., Trottier Y., Lehrach H., Davies S.W. and Bates G.P. (1996) Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell. 87:493-506]. Transgenic mice were treated with rolipram 1.5 mg/kg daily starting from 4 weeks of age. After transcardial perfusion, histological and immunohistochemical studies were performed. We found that rolipram-treated R6/2 mice survived longer and displayed less severe signs of neurological dysfunction than the vehicle treated ones. Primary outcome measures such as brain volume, striatal atrophy, size and morphology of striatal neurons, neuronal intranuclear inclusions and microglial reaction confirmed a neuroprotective effect of the compound. Rolipram was effective in increasing significantly the levels of activated CREB and of BDNF the striatal spiny neurons, which might account for the beneficial effects observed in this model. Our findings show that rolipram could be considered as a valid therapeutic approach for HD.
Collapse
|
220
|
Kotz CM, Teske JA, Billington CJ. Neuroregulation of nonexercise activity thermogenesis and obesity resistance. Am J Physiol Regul Integr Comp Physiol 2008; 294:R699-710. [PMID: 18160530 DOI: 10.1152/ajpregu.00095.2007] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
High levels of spontaneous physical activity in lean people and the nonexercise activity thermogenesis (NEAT) derived from that activity appear to protect lean people from obesity during caloric challenge, while obesity in humans is characterized by dramatically reduced spontaneous physical activity. We have similarly demonstrated that obesity-resistant rats have significantly greater spontaneous physical activity than obesity-prone rats, and that spontaneous physical activity predicts body weight gain. Although the energetic cost of activity varies between types of activity and may be regulated, individual level of spontaneous physical activity is important in determining propensity for obesity. We review the current status of knowledge about the brain mechanisms involved in controlling the level of spontaneous physical activity and the NEAT so generated. Focus is on potential neural mediators of spontaneous physical activity and NEAT, including orexin A (also known as hypocretin 1), agouti-related protein, ghrelin, and neuromedin U, in addition to brief mention of neuropeptide Y, corticotrophin releasing hormone, cholecystokinin, estrogen, leptin, and dopamine effects on spontaneous physical activity. We further review evidence that strain differences in orexin stimulation pathways for spontaneous physical activity and NEAT appear to track with the body weight phenotype, thus providing a potential mechanistic explanation for reduced activity and weight gain.
Collapse
Affiliation(s)
- Catherine M Kotz
- Veterans Affairs Medical Center, GRECC (11G) One Veterans Drive, Minneapolis, MN 55417, USA.
| | | | | |
Collapse
|
221
|
Rosa MPD, Baroni GV, Portal VL. Contribuição na prevenção da isquemia cerebral pelo cilostazol, um inibidor da fosfodiesterase III: revisão de literatura. J Vasc Bras 2008. [DOI: 10.1590/s1677-54492008000100009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Esta revisão bibliográfica objetiva expor estas pesquisas sobre as ações do cilostazol no sistema nervoso central. O cilostazol é uma droga que demonstrou exercer inibição seletiva e potente da fosfodiesterase tipo III, ocasionando o aumento de adenosina cíclica -3',5'-monofosfato nas plaquetas, nas células endoteliais e nas células musculares lisas, sendo classificado como vasodilatador, antiagregante plaquetário e antitrombótico. É o fármaco de primeira escolha na claudicação intermitente devido à doença arterial obstrutiva periférica. Além disso, há evidências de que o cilostazol é eficaz no processo aterosclerótico cerebral, promovendo aumento do fluxo e volume sangüíneos e prevenindo infartos, especialmente lacunares e recorrentes, por diminuir a morte celular devido à apoptose e ao estresse oxidativo nas substâncias branca e parda.
Collapse
|
222
|
O’Neill C. The potential roles for embryotrophic ligands in preimplantation embryo development. Hum Reprod Update 2008; 14:275-88. [DOI: 10.1093/humupd/dmn002] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
223
|
Dwivedi Y, Pandey GN. Adenylyl cyclase-cyclicAMP signaling in mood disorders: role of the crucial phosphorylating enzyme protein kinase A. Neuropsychiatr Dis Treat 2008; 4:161-76. [PMID: 18728821 PMCID: PMC2515915 DOI: 10.2147/ndt.s2380] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mood disorders are among the most prevalent and recurrent forms of psychiatric illnesses. In the last decade, there has been increased understanding of the biological basis of mood disorders. In fact, novel mechanistic concepts of the neurobiology of unipolar and bipolar disorders are evolving based on recent pre-clinical and clinical studies, most of which now focus on the role of signal transduction mechanisms in these psychiatric illnesses. Particular investigative emphasis has been given to the role of phosphorylating enzymes, which are crucial in regulating gene expression and neuronal and synaptic plasticity. Among the most important phosphorylating enzyme is protein kinase A (PKA), a component of adenylyl cyclase-cyclic adenosine monophosphate (AC-cAMP) signaling system. In this review, we critically and comprehensively discuss the role of various components of AC-cAMP signaling in mood disorders, with a special focus on PKA, because of the interesting observation that have been made about its involvement in unipolar and bipolar disorders. We also discuss the functional significance of the findings regarding PKA by discussing the role of important PKA substrates, namely, Rap-1, cyclicAMP-response element binding protein, and brain-derived neurotrophic factor. These studies suggest the interesting possibility that PKA and related signaling molecules may serve as important neurobiological factors in mood disorders and may be relevant in target-specific therapeutic interventions for these disorders.
Collapse
Affiliation(s)
- Yogesh Dwivedi
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago Chicago, Illinois 60612, USA.
| | | |
Collapse
|
224
|
Marini AM, Jiang H, Pan H, Wu X, Lipsky RH. Hormesis: a promising strategy to sustain endogenous neuronal survival pathways against neurodegenerative disorders. Ageing Res Rev 2008; 7:21-33. [PMID: 17889623 DOI: 10.1016/j.arr.2007.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 07/31/2007] [Accepted: 07/31/2007] [Indexed: 12/22/2022]
Abstract
The brain developed adaptive mechanisms in the face of changing environments and stresses imposed on the nervous system. The addition of glutamate as the major excitatory amino acid neurotransmitter to the brain's complement of amino acids and peptides dictated a coordinated transcriptional and translational program to meet the demands of excitatory neurotransmission. One such program is the ability of neurons to sustain and maintain their survival given the nature of glutamate-mediated receptor activation. The unique development of endogenous neuronal pathways activated by glutamate receptors transformed neurons and allowed them to survive under conditions of high energy demands. These same endogenous survival pathways also mediate plastic responses to meet another demand of the brain, adaptation. An endogenous protein that plays a central role in glutamate receptor-mediated survival pathways is brain-derived neurotrophic factor (BDNF). Intermittent but frequent synaptic ionotropic glutamate receptor activation ensures neuronal survival through a BDNF autocrine loop. In sharp contrast, overactivation of ionotropic glutamate receptors leads to neuronal cell death. Thus, innovative strategies that induce endogenous neuronal survival pathways through low-level activation of ionotropic glutamate receptors or those that bypass receptor activation but upregulate endogenous survival pathways may not only prevent neurodegenerative disorders that involve glutamate as a final common pathway that kills neurons, but may also provide treatment alternatives critical for neurons to survive stressful conditions such as stroke, status epilepticus and hypoglycemic-induced neuronal cell death.
Collapse
Affiliation(s)
- Ann M Marini
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | | | | | | | | |
Collapse
|
225
|
Sawe N, Steinberg G, Zhao H. Dual roles of the MAPK/ERK1/2 cell signaling pathway after stroke. J Neurosci Res 2008; 86:1659-69. [DOI: 10.1002/jnr.21604] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
226
|
Ischemic tolerance as an active and intrinsic neuroprotective mechanism. HANDBOOK OF CLINICAL NEUROLOGY 2008; 92:171-95. [PMID: 18790275 DOI: 10.1016/s0072-9752(08)01909-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
227
|
O'Driscoll C, O'Connor J, O'Brien CJ, Cotter TG. Basic fibroblast growth factor-induced protection from light damage in the mouse retina in vivo. J Neurochem 2007; 105:524-36. [PMID: 18088352 DOI: 10.1111/j.1471-4159.2007.05189.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Basic fibroblast growth factor (bFGF) has proven neuroprotective efficacy in the rodent retina against a diverse array of injurious stimuli. However, there is no consensus to date as to the molecular mechanisms underlying this neuroprotection. The study presented herein demonstrates increased expression of endogenous bFGF in the albino mouse retina in response to acute exposure to sublethal levels of light stress. The increased expression correlates with significant photoreceptor protection from light damage. The neuroprotection is likely to be mediated by bFGF as we demonstrate that a shorter exposure to bright light stress that does not up-regulate bFGF fails to protect photoreceptors from light damage. Furthermore, intravitreal bFGF injection into the retina of mice 3 h prior to light damage affords almost complete photoreceptor protection from light-induced degeneration. In addition, injected bFGF induces the activation of protein kinase B and extracellular signal-regulated kinase 1/2 signalling which correlate directly with the pathways we find to be activated in response to light stress and up-regulated bFGF. Moreover, we demonstrate that both bright light pre-conditioning and intravitreal bFGF injection result in dramatic increases in levels of inactive glycogen synthase kinase 3beta and cyclic AMP response element binding protein phosphorylation indicating a potential mechanism by which bFGF promotes survival of photoreceptors in vivo.
Collapse
Affiliation(s)
- Carolyn O'Driscoll
- Cell Development and Disease Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
228
|
Abstract
The N-methyl-D-aspartate (NMDA) subtype of ionotropic glutamate receptors plays a Jekyll and Hyde role in the mammalian central nervous system. In pathological scenarios such as ischemia, Ca2+ influx through the NMDA receptor is a key mediator of cell death. However, physiological levels of NMDA-receptor activity can promote neuronal survival and resistance to trauma and play important roles in synaptic plasticity and transmission. This dichotomy may explain the poor tolerance and efficacy of NMDA-receptor antagonists in clinical trials for excitotoxic trauma. There is a growing understanding of the signaling events that mediate the opposing effects of NMDA-receptor activity and the factors that determine whether an episode of NMDA-receptor activity will promote survival or death. This knowledge may lead to therapeutic strategies that enable the selective blockade of prodeath signaling cassettes while sparing physiological signaling to survival and plasticity.
Collapse
Affiliation(s)
- Sofia Papadia
- Centre for Neuroscience Research, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Giles E. Hardingham
- Centre for Neuroscience Research, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| |
Collapse
|
229
|
Barrera LO, Li Z, Smith AD, Arden KC, Cavenee WK, Zhang MQ, Green RD, Ren B. Genome-wide mapping and analysis of active promoters in mouse embryonic stem cells and adult organs. Genome Res 2007; 18:46-59. [PMID: 18042645 DOI: 10.1101/gr.6654808] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
By integrating genome-wide maps of RNA polymerase II (Polr2a) binding with gene expression data and H3ac and H3K4me3 profiles, we characterized promoters with enriched activity in mouse embryonic stem cells (mES) as well as adult brain, heart, kidney, and liver. We identified approximately 24,000 promoters across these samples, including 16,976 annotated mRNA 5' ends and 5153 additional sites validating cap-analysis of gene expression (CAGE) 5' end data. We showed that promoters with CpG islands are typically non-tissue specific, with the majority associated with Polr2a and the active chromatin modifications in nearly all the tissues examined. By contrast, the promoters without CpG islands are generally associated with Polr2a and the active chromatin marks in a tissue-dependent way. We defined 4396 tissue-specific promoters by adapting a quantitative index of tissue-specificity based on Polr2a occupancy. While there is a general correspondence between Polr2a occupancy and active chromatin modifications at the tissue-specific promoters, a subset of them appear to be persistently marked by active chromatin modifications in the absence of detectable Polr2a binding, highlighting the complexity of the functional relationship between chromatin modification and gene expression. Our results provide a resource for exploring promoter Polr2a binding and epigenetic states across pluripotent and differentiated cell types in mammals.
Collapse
Affiliation(s)
- Leah O Barrera
- Ludwig Institute for Cancer Research, UCSD School of Medicine, La Jolla, California 92093-0653, USA
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Fogal B, Li J, Lobner D, McCullough LD, Hewett SJ. System x(c)- activity and astrocytes are necessary for interleukin-1 beta-mediated hypoxic neuronal injury. J Neurosci 2007; 27:10094-105. [PMID: 17881516 PMCID: PMC6672668 DOI: 10.1523/jneurosci.2459-07.2007] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The purpose of this study was to elucidate the cellular/biochemical pathway(s) by which interleukin-1beta (IL-1beta) contributes to the pathogenesis of hypoxic-ischemic brain damage. In vivo, IL-1 receptor type I (IL-1RI)-deficient mice showed smaller infarcts and less neurological deficits than wild-type animals after a 90 min reversible middle cerebral artery occlusion. In vitro, IL-1beta mediated an enhancement of hypoxic neuronal injury in murine cortical cultures that was lacking in cultures derived from IL-1RI null mutant animals and was blocked by the IL-1 receptor antagonist or an IL-1RI blocking antibody. This IL-1beta-mediated potentiation of hypoxic neuronal injury was associated with an increase in both cellular cystine uptake ([cystine]i) and extracellular glutamate levels ([glutamate]e) and was prevented by either ionotropic glutamate receptor antagonism or removal of L-cystine, suggesting a role for the cystine/glutamate antiporter (System x(c)-). Indeed, dual System x(c)-/metabotropic glutamate receptor subunit 1 (mGluR1) antagonism but not selective mGluR1 antagonism prevented neuronal injury. Additionally, cultures derived from mGluR1-deficient mice exhibited the same potentiation in injury after treatment with IL-1beta as wild-type cultures, an effect prevented by System x(c)-/mGluR1 antagonism. Finally, assessment of System x(c)- function and kinetics in IL-1beta-treated cultures revealed an increase in velocity of cystine transport (Vmax), in the absence of a change in affinity (Km). Neither the enhancement in [cystine]i, [glutamate]e, or neuronal injury were observed in chimeric cultures consisting of IL-1RI(+/+) neurons plated on top of IL-1RI(-/-) astrocytes, highlighting the importance of astrocyte-mediated alterations in System x(c)- as a novel contributor to the development and progression of hypoxic neuronal injury.
Collapse
Affiliation(s)
| | - Jun Li
- Neurology, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| | - Doug Lobner
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53233
| | - Louise D. McCullough
- Neurology, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| | | |
Collapse
|
231
|
Lin X, Zhang Y, Dong J, Zhu X, Ye M, Shi J, Lu J, Di Q, Shi J, Liu W. GM-CSF enhances neural differentiation of bone marrow stromal cells. Neuroreport 2007; 18:1113-7. [PMID: 17589309 DOI: 10.1097/wnr.0b013e3282010aff] [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] [Indexed: 11/26/2022]
Abstract
Recent reports suggest that bone marrow stromal cells may be induced into neural cells both in vivo and in vitro. The factors that regulate the neural differentiation and the mechanism involved, however, remains unclear. Here we demonstrated that granulocyte-macrophage colony-stimulating factor (GM-CSF), a potent hematopoietic factor, was able to enhance the neural differentiation of bone marrow stromal cells. Moreover, we found that GM-CSF receptors are abundantly distributed in the bone marrow stromal cells and GM-CSF significantly upregulated the phosphorylation of cAMP-responsive element binding protein in bone marrow stromal cells. These findings suggest that GM-CSF may activate its receptor and then enhance neural differentiation of bone marrow stromal cells by upregulating phosphorylation of cAMP-responsive element binding protein.
Collapse
Affiliation(s)
- Xingjian Lin
- Department of Neurology, Nanjing Medical University, Nanjing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Pandey GN, Dwivedi Y, Ren X, Rizavi HS, Roberts RC, Conley RR. Cyclic AMP response element-binding protein in post-mortem brain of teenage suicide victims: specific decrease in the prefrontal cortex but not the hippocampus. Int J Neuropsychopharmacol 2007; 10:621-9. [PMID: 16978443 DOI: 10.1017/s1461145706007231] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Abnormalities in both adenylyl cyclase (AC) and phosphoinositide (PI) signalling systems have been observed in the post-mortem brain of suicide victims. Cyclic AMP response element-binding protein (CREB) is a transcription factor that is activated by phosphorylating enzymes such as protein kinase A (PKA) and protein kinase C (PKC), which suggests that both AC and PI signalling systems converge at the level of CREB. CREB is involved in the transcription of many neuronally expressed genes that have been implicated in the pathophysiology of depression and suicide. Since we observed abnormalities of both PKA and PKC in the post-mortem brain of teenage suicide victims, we examined if these abnormalities are also associated with abnormalities of CREB, which is activated by these phosphorylating enzymes. We determined CRE-DNA binding using the gel shift assay, as well as protein expression of CREB using the Western blot technique, and the mRNA expression of CREB using a quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) technique in the prefrontal cortex (PFC), and hippocampus obtained from 17 teenage suicide victims and 17 matched normal control subjects. We observed that the CRE-DNA binding and the protein expression of CREB were significantly decreased in the PFC of teenage suicide victims compared with controls. There was also a significant decrease in mRNA expression of CREB in the PFC of teenage suicide victims compared with control subjects. However, there were no significant differences in CRE-DNA binding or the protein and mRNA expression of CREB in the hippocampus of teenage suicide victims compared with control subjects. These results suggest that the abnormalities of PKA, and of PKC, observed in teenage suicide victims are also associated with abnormalities of the transcription factor CREB, and that this may also cause alterations of important neuronally expressed genes, and provide further support of the signal transduction of abnormalities in suicide.
Collapse
Affiliation(s)
- Ghanshyam N Pandey
- The Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | | | | | | | | | | |
Collapse
|
233
|
Takeda H, Kitaoka Y, Hayashi Y, Kumai T, Munemasa Y, Fujino H, Kobayashi S, Ueno S. Calcium/calmodulin-dependent protein kinase II regulates the phosphorylation of CREB in NMDA-induced retinal neurotoxicity. Brain Res 2007; 1184:306-15. [PMID: 17961520 DOI: 10.1016/j.brainres.2007.09.055] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2007] [Revised: 09/17/2007] [Accepted: 09/17/2007] [Indexed: 01/15/2023]
Abstract
We examined the role of the phosphorylation of calcium/calmodulin-dependent protein kinase II (CaMKII) and cyclic AMP-response element binding protein (CREB) in N-methyl-d-aspartate (NMDA)-induced neurotoxicity in the rat retina. Western blot analysis showed early elevation of phosphorylated CaMKII (p-CaMKII) protein levels and subsequential elevation of phosphorylated CREB (p-CREB) protein after NMDA injection. Immunohistochemistry showed that p-CaMKII was colocalized with Thy-1-positive retinal ganglion cells (RGCs) after NMDA injection. The increase in the p-CaMKII protein level was significantly inhibited by the preinjection of CaMKII small interfering RNA (siRNA), whereas negative control siRNA did not affect. Moreover, the increase in the p-CREB protein level after NMDA injection was also prevented by preinjection of CaMKII siRNA. In addition, our morphometric study of neurotracer retrograde labeling and Thy-1-positive cells showed that CaMKII siRNA significantly accelerated NMDA-induced RGC loss. Furthermore, the prevention of CREB binding by CRE decoy oligonucleotide also exacerbated RGC loss. These results suggest that the activation of CaMKII may regulate CREB phosphorylation and that the transient phosphorylation of CaMKII and CREB may be a neuroprotective response against NMDA-induced neurotoxicity.
Collapse
Affiliation(s)
- Hiroyuki Takeda
- Department of Ophthalmology, St Marianna University School of Medicine, Kawasaki-Shi, Kanagawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
234
|
Weng Z, Signore AP, Gao Y, Wang S, Zhang F, Hastings T, Yin XM, Chen J. Leptin protects against 6-hydroxydopamine-induced dopaminergic cell death via mitogen-activated protein kinase signaling. J Biol Chem 2007; 282:34479-91. [PMID: 17895242 DOI: 10.1074/jbc.m705426200] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The death of midbrain dopaminergic neurons in sporadic Parkinson disease is of unknown etiology but may involve altered growth factor signaling. The present study showed that leptin, a centrally acting hormone secreted by adipocytes, rescued dopaminergic neurons, reversed behavioral asymmetry, and restored striatal catecholamine levels in the unilateral 6-hydroxydopamine (6-OHDA) mouse model of dopaminergic cell death. In vitro studies using the murine dopaminergic cell line MN9D showed that leptin attenuated 6-OHDA-induced apoptotic markers, including caspase-9 and caspase-3 activation, internucleosomal DNA fragmentation, and cytochrome c release. ERK1/2 phosphorylation (pERK1/2) was found to be critical for mediating leptin-induced neuroprotection, because inhibition of the MEK pathway blocked both the pERK1/2 response and the pro-survival effect of leptin in cultures. Knockdown of the downstream messengers JAK2 or GRB2 precluded leptin-induced pERK1/2 activation and neuroprotection. Leptin/pERK1/2 signaling involved phosphorylation and nuclear localization of CREB (pCREB), a well known survival factor for dopaminergic neurons. Leptin induced a marked MEK-dependent increase in pCREB that was essential for neuroprotection following 6-OHDA toxicity. Transfection of a dominant negative MEK protein abolished leptin-enhanced pCREB formation, whereas a dominant negative CREB or decoy oligonucleotide diminished both pCREB binding to its target DNA sequence and MN9D survival against 6-OHDA toxicity. Moreover, in the substantia nigra of mice, leptin treatment increased the levels of pERK1/2, pCREB, and the downstream gene product BDNF, which were reversed by the MEK inhibitor PD98059. Collectively, these data provide evidence that leptin prevents the degeneration of dopaminergic neurons by 6-OHDA and may prove useful in the treatment of Parkinson disease.
Collapse
Affiliation(s)
- Zhongfang Weng
- Department of Neurology, and Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
235
|
D-serine regulates CREB phosphorylation induced by NMDA receptor activation in Müller glia from the retina. Neurosci Lett 2007; 427:55-60. [PMID: 17920195 DOI: 10.1016/j.neulet.2007.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 08/28/2007] [Accepted: 09/09/2007] [Indexed: 11/23/2022]
Abstract
D-serine is an N-methyl-D-aspartate (NMDA) receptor coagonist predominantly produced by glial cells in the brain and the retina. Whereas a role for D-serine as a modulator of NMDA receptors in neurons has been suggested, its function in glial cells has not been analyzed. We here show that D-serine modulates gene expression in Müller glial cells from the retina through the induction of transcription factor CREB phosphorylation and the expression of the immediate-early gene c-fos. Pharmacological analysis indicates that D-serine effect involves NMDA receptor activation. Comparison of the effect of D-serine in Müller cells, hippocampal astrocytes and hippocampal neurons suggests that D-serine could function as a retinal NMDA receptor coagonist activating functionally relevant transcription factor pathways in glial cells.
Collapse
|
236
|
Micheli V, Sestini S, Parri V, Fichera M, Romano C, Ariani F, Longo I, Mari F, Bruttini M, Renieri A, Meloni I. RSK2 enzymatic assay as a second level diagnostic tool in Coffin-Lowry syndrome. Clin Chim Acta 2007; 384:35-40. [PMID: 17586481 DOI: 10.1016/j.cca.2007.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 04/17/2007] [Accepted: 05/14/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND Coffin-Lowry syndrome is a semi-dominant condition characterized by severe psychomotor retardation with facial, hand and skeletal malformations resulting from mutations in RSK2 gene, encoding for a serine/threonine kinase. More than 100 different mutations have been identified to date; however, about 50% of clinically diagnosed patients apparently do not have mutations. In order to exclude that these patients have RSK2 mutations missed by standard mutation detection techniques, a rapid and sensitive assay allowing evaluation of RSK2 activity was needed. METHODS RSK2 capacity to phosphorylate a synthetic CREB-peptide in basal and PMA-stimulated conditions was evaluated in lymphoblasts from 3 patients with RSK2 mutations and normal controls. RESULTS Patients RSK2 activity is normal in nonstimulated conditions but fails to grow following stimulation. The evaluation of the stimulated/non-stimulated activity ratio demonstrated a statistically significant impairment in patients. CONCLUSIONS We have set up an assay which allows the identification of even partial alterations of RSK2 activity and seems to give good results also in females with a balanced X-chromosome inactivation and thus with a presumably normal enzymatic activity in about 50% of cells. Moreover, our data seem to confirm previous reports of a potential direct correlation between the level of RSK2 activity and the severity of cognitive impairment.
Collapse
Affiliation(s)
- Vanna Micheli
- Biological Chemistry, Department of Molecular Biology, University of Siena, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Rodríguez A, Ferrer I. Expression of transcription factors CREB and c-Fos in the brains of terminal Creutzfeldt-Jakob disease cases. Neurosci Lett 2007; 421:10-5. [PMID: 17548164 DOI: 10.1016/j.neulet.2007.04.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 03/22/2007] [Accepted: 04/06/2007] [Indexed: 10/23/2022]
Abstract
Expression levels and localization of transcription factors cAMP response element binding protein (CREB(1) and CREB(2)) and c-Fos, as well as levels of up-stream mitogen-activated protein kinases/extracellular signal-regulated kinases (ERK-1 and ERK-2) and p38 kinase, were examined in the brains (frontal cortex) of eleven cases with Creutzfeldt-Jakob disease (CJD) and five age-matched controls. Preserved expression levels of ERK-1-P, ERK-2-P and p38-P were observed in CJD. However, significantly reduced levels, as revealed by gel electrophoresis and Western blotting, and reduced numbers of immunoreactive nuclei, as seen by immunohistochemistry, to CREB, CREB-P, c-Fos and c-Fos-P were found in CJD when compared with controls. These observations point to exhausted CREB and c-Fos brain responses, in spite of preserved up-stream signaling kinases, thus favoring cell death in terminal stages of CJD.
Collapse
Affiliation(s)
- A Rodríguez
- Institut de Neuropatologia, Servei Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | | |
Collapse
|
238
|
Schölzke MN, Schwaninger M. Transcriptional regulation of neurogenesis: potential mechanisms in cerebral ischemia. J Mol Med (Berl) 2007; 85:577-88. [PMID: 17429598 DOI: 10.1007/s00109-007-0196-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Revised: 02/27/2007] [Accepted: 03/20/2007] [Indexed: 12/11/2022]
Abstract
Recent data provides evidence that new neurons are born in cerebral ischemia. Although ultimate evidence for their functional importance is lacking, correlational data suggest that they contribute to recovery. Therefore, the underlying mechanisms of neurogenesis are interesting as a basis for pharmacological enhancement of the phenomenon. Neurogenesis is a multistep process that includes proliferation of precursor cells, migration of the newborn cells to the site of lesion, differentiation, integration into neuronal circuits, and survival. All these steps rely on gene transcription. However, only preliminary data about the specific transcriptional control of neurogenesis in cerebral ischemia have been obtained so far. To promote this investigation, we review currently available information on six pathways (Notch, Wnt/beta-catenin, NF-kappaB, signal transducers and activators of transcription (STA) 3, HIF-1, and cyclic AMP response element-binding protein [CREB]) that have been shown to regulate transcription in neurogenesis and that have been implicated in cerebral ischemia. With the exception of CREB, direct involvement in postischemic neurogenesis is quite conjectural and much more must be learned to draw practical conclusions.
Collapse
Affiliation(s)
- Marion N Schölzke
- Department of Neurology, University of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | | |
Collapse
|
239
|
Kim DW, Lee JH, Park SK, Yang WM, Jeon GS, Lee YH, Chung CK, Cho SS. Astrocytic Expressions of Phosphorylated Akt, GSK3β and CREB Following an Excitotoxic Lesion in the Mouse Hippocampus. Neurochem Res 2007; 32:1460-8. [PMID: 17417726 DOI: 10.1007/s11064-007-9332-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Accepted: 03/15/2007] [Indexed: 01/07/2023]
Abstract
Glycogen synthase kinase 3beta (GSK3beta) is believed to play important roles in the regulation of synaptic plasticity, cell survival and circadian rhythms in the mature CNS. However, although several studies have been focused on the GSK3beta, little is known about GSK3beta changes in glial cells under neuropathological conditions. In this study, we evaluated the expressions of molecules associated with the GSK3beta signaling pathway, following the induction of an excitotoxic lesion in mouse brain by kainic acid (KA) injection, which caused pyramidal cell degeneration in the hippocampal CA3 region. In injured hippocampi, Ser47-Akt (protein kinase B, PKB) phosphorylation increased from 4 h until 1 day post-injection (PI). Ser9-GSK3beta and Ser133-cAMP responsive element-binding protein (CREB) phosphorylations showed similar spatiotemporal patterns in hippocampi at 1 day until 3 days PI. Double immunohistochemistry also showed that these phosphorylated forms of Akt, GSK3beta and CREB were expressed in astrocytes. For the first time, our data demonstrate the injury-induced astrocytic changes in the levels of phosphorylation of Akt, -GSK3beta and -CREB in vivo, which may reflect mechanisms of glial cells protection or adaptive response to damage.
Collapse
Affiliation(s)
- Dong Woon Kim
- Department of Anatomy, Chungnam National University College of Medicine, Daejeon 301-131, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
240
|
Sönmez A, Kabakçi B, Vardar E, Gürel D, Sönmez U, Orhan YT, Açikel U, Gökmen N. Erythropoietin attenuates neuronal injury and potentiates the expression of pCREB in anterior horn after transient spinal cord ischemia in rats. ACTA ACUST UNITED AC 2007; 68:297-303; discussion 303. [PMID: 17368520 DOI: 10.1016/j.surneu.2006.11.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Accepted: 11/03/2006] [Indexed: 11/29/2022]
Abstract
BACKGROUND Recent studies have suggested that EPO activates the CREB transcription pathway and increases BDNF expression and production, which contributes to EPO-mediated neuroprotection. We investigated whether EPO has a neuroprotective effect against ISCI in rats and examined the involvement of CREB protein phosphorylation in this process. METHODS Spinal cord ischemia was produced by balloon occlusion of the abdominal aorta below the branching point of the left subclavian artery for 5 minutes, and rHu-EPO (1000 U/kg BW) was administered intravenously after the onset of the reperfusion. Neurologic status was assessed at 1, 24, and, 48 hours. After the end of 48 hours, spinal cords were harvested for histopathologic analysis and immunohistochemistry for pCREB. RESULTS All sham-operated rats had a normal neurologic outcome, whereas all ischemic rats suffered severe neurologic deficits after ISCI. Erythropoietin treatment was found to accelerate recovery of motor deficits and prevent the loss of motoneurons in the spinal cord after transient ischemia. Ischemic spinal cord injury induced the phosphorylation of pCREB at the anterior horn of the spinal cord, and EPO treatment significantly potentiated expression of pCREB increase at the anterior horn of the spinal cord. CONCLUSIONS These results demonstrate that a single dose of EPO given before ISCI provides significant neuroprotection and potentiates the expression of pCREB in this region.
Collapse
Affiliation(s)
- Ataç Sönmez
- Learning Resources Center Research Laboratory, School of Medicine, Dokuz Eylul University Inciralti, TR-35340, Izmir, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
241
|
Lin YL, Lee YC, Huang CL, Lai WL, Lin YR, Huang NK. Ligusticum chuanxiong prevents rat pheochromocytoma cells from serum deprivation-induced apoptosis through a protein kinase A-dependent pathway. JOURNAL OF ETHNOPHARMACOLOGY 2007; 109:428-34. [PMID: 16973320 DOI: 10.1016/j.jep.2006.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 07/11/2006] [Accepted: 08/11/2006] [Indexed: 05/11/2023]
Abstract
Ligusticum chuanxiong (LC) is a traditional Chinese herbal medicine used to treat various cardiovascular diseases. In this study, the butanol extract of LC was found to protect neuronal-like pheochromocytoma cells from serum deprivation-induced apoptosis. Both a serine/threonine kinase inhibitor and a specific protein kinase A (PKA) inhibitor blocked the protective effect of LC. A transcription inhibitor (actinomycin D) and a protein synthesis inhibitor (cyclohexamide) also attenuated the protective effect of LC, suggesting the requirement of gene expression for the protection of LC. On the other hand, LC increased both the formation of cyclic-AMP and the phosphorylation of the cyclic-AMP response element-binding protein (CREB), a downstream target of PKA and a nuclear transcription factor known for neuroprotective mechanism. Furthermore, LC-induced CREB phosphorylation and protective effect could be blocked by a PKA inhibitor and overexpression of the dominant negative CREB, respectively. Taken together, the protective mechanism of LC in antagonizing serum deprivation-induced PC12 cell apoptosis might be mediated through a PKA/CREB-dependent pathway.
Collapse
Affiliation(s)
- Yun-Lian Lin
- National Research Institute of Chinese Medicine, No. 155-1, Li-Nung Street, Section 2, Shipai, Peitou, Taipei 112, Taiwan
| | | | | | | | | | | |
Collapse
|
242
|
Sahin M, Saxena A, Joost P, Lewerenz J, Methner A. Induction of Bcl-2 by functional regulation of G-protein coupled receptors protects from oxidative glutamate toxicity by increasing glutathione. Free Radic Res 2007; 40:1113-23. [PMID: 17050165 DOI: 10.1080/10715760600838191] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Glutamate treatment depletes hippocampal HT22 cells of glutathione, which renders the cells incapable to reduce reactive oxygen species and ultimately cumulates in cell death by oxidative stress. HT22 cells resistant to glutamate displayed increased phosphorylation of cAMP-response-element binding (CREB) and decreased ERK1/2 suggestive of differences in signal transmission. We investigated the amount of candidate G-protein-coupled receptors involved in this resistance and found an increase in mRNA for receptors activated by the vasoactive intestinal peptide VIP (VPAC2, 12.6-fold) and glutamate like the metabotropic glutamate receptor mGlu1 (5.3-fold). Treating cells with VIP and glutamate led to the same changes in protein phosphorylation observed in resistant cells and induced the proto-oncogene Bcl-2. Bcl-2 overexpression protected by increasing the amount of intracellular glutathione and Bcl-2 knockdown by small interfering RNAs (siRNA) increased glutamate susceptibility of resistant cells. Other receptors upregulated in this paradigm might represent useful targets in the treatment of neurological diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Mert Sahin
- Department of Neurology, Heinrich Heine Universität Düsseldorf, Moorenstreet 5, 40225, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
243
|
Zhang SJ, Steijaert MN, Lau D, Schütz G, Delucinge-Vivier C, Descombes P, Bading H. Decoding NMDA Receptor Signaling: Identification of Genomic Programs Specifying Neuronal Survival and Death. Neuron 2007; 53:549-62. [PMID: 17296556 DOI: 10.1016/j.neuron.2007.01.025] [Citation(s) in RCA: 250] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Revised: 11/15/2006] [Accepted: 01/26/2007] [Indexed: 12/16/2022]
Abstract
NMDA receptors promote neuronal survival but also cause cell degeneration and neuron loss. The mechanisms underlying these opposite effects on neuronal fate are unknown. Whole-genome expression profiling revealed that NMDA receptor signaling is decoded at the genomic level through activation of two distinct, largely nonoverlapping gene-expression programs. The location of the NMDA receptor activated specifies the transcriptional response: synaptic NMDA receptors induce a coordinate upregulation of newly identified pro-survival genes and downregulation of pro-death genes. Extrasynaptic NMDA receptors fail to activate this neuroprotective program, but instead induce expression of Clca1, a putative calcium-activated chloride channel that kills neurons. These results help explain the opposing roles of synaptic and extrasynaptic NMDA receptors on neuronal fate. They also demonstrate that the survival function is implemented in neurons through a multicomponent system of functionally related genes, whose coordinate expression is controlled by specific calcium signal initiation sites.
Collapse
Affiliation(s)
- Sheng-Jia Zhang
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
244
|
Cerezo-Guisado M, GarcíA-Román N, García-MaríN L, Álvarez-Barrientos A, Bragado M, Lorenzo M. Lovastatin inhibits the extracellular-signal-regulated kinase pathway in immortalized rat brain neuroblasts. Biochem J 2007; 401:175-83. [PMID: 16952276 PMCID: PMC1698684 DOI: 10.1042/bj20060731] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have shown previously that lovastatin, a 3-hydroxy-3-methyl- glutaryl coenzyme A reductase inhibitor, induces apoptosis in spontaneously immortalized rat brain neuroblasts. In the present study, we analysed the intracellular signal transduction pathways by which lovastatin induces neuroblast apoptosis. We showed that lovastatin efficiently inhibited Ras activation, which was associated with a significant decrease in ERK1/2 (extracellular-signal-regulated kinase 1/2) phosphorylation. Lovastatin also decreased CREB phosphorylation and CREB-mediated gene expression. The effects of lovastatin on the Ras/ERK1/2/CREB pathway were time- and concentration-dependent and fully prevented by mevalonate. In addition, we showed that two MEK [MAPK (mitogen-activated protein kinase)/ERK kinase] inhibitors, PD98059 and PD184352, were poor inducers of apoptosis in serum-treated neuroblasts. However, these inhibitors significantly increased apoptosis induced by lovastatin treatment. Furthermore, we showed that pharmacological inhibition of both MEK and phosphoinositide 3-kinase activities was able to induce neuroblast apoptosis with similar efficacy as lovastatin. Our results suggest that lovastatin triggers neuroblast apoptosis by regulating several signalling pathways, including the Ras/ERK1/2 pathway. These findings might also contribute to elucidate the intracellular mechanisms involved in the central nervous system side effects associated with statin therapy.
Collapse
Affiliation(s)
- Maria Isabel Cerezo-Guisado
- *Departamento de Bioquímica, Biología Molecular y Genética Universidad de Extremadura, E-10071 Cáceres, Spain
| | - Natalia GarcíA-Román
- †Departamento de Bioquímica y Biología Molecular, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| | | | | | - Maria Julia Bragado
- *Departamento de Bioquímica, Biología Molecular y Genética Universidad de Extremadura, E-10071 Cáceres, Spain
| | - Maria Jesús Lorenzo
- *Departamento de Bioquímica, Biología Molecular y Genética Universidad de Extremadura, E-10071 Cáceres, Spain
- To whom correspondence should be addressed (email )
| |
Collapse
|
245
|
Karaçay B, Li G, Pantazis NJ, Bonthius DJ. Stimulation of the cAMP pathway protects cultured cerebellar granule neurons against alcohol-induced cell death by activating the neuronal nitric oxide synthase (nNOS) gene. Brain Res 2007; 1143:34-45. [PMID: 17306238 PMCID: PMC1894830 DOI: 10.1016/j.brainres.2007.01.059] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2006] [Revised: 01/12/2007] [Accepted: 01/15/2007] [Indexed: 12/01/2022]
Abstract
Neuronal loss is a key component of fetal alcohol syndrome pathophysiology. Therefore, identification of molecules and signaling pathways that ameliorate alcohol-induced neuronal death is important. We have previously reported that neuronal nitric oxide synthase (nNOS) can protect developing cerebellar granule neurons (CGN) against alcohol-induced death both in vitro and in vivo. However, the upstream signal controlling nNOS expression in CGN is unknown. Activated cAMP response element binding protein (CREB) has been strongly linked to the survival of multiple cell types, including CGN. Furthermore, the promoter of the nNOS gene contains two cAMP response elements (CRE). Using cultures of CGN, we tested the hypothesis that cAMP mediates nNOS activation and the protective effect of nNOS against alcohol-induced cell death. Forskolin, an activator of the cAMP pathway, stimulated expression of a reporter gene under the control of the nNOS promoter, and this stimulation was substantially reduced when the two CREs were mutated. Forskolin increased nNOS mRNA levels several fold, increased production of nitric oxide, and abolished alcohol's toxic effect in wild type CGN. Furthermore, forskolin's protective effect was substantially reduced in CGN cultures genetically deficient for nNOS (from nNOS-/- mice). Delivery of nNOS cDNA using a replication-deficient adenoviral vector into nNOS-/- CGN abolished alcohol-induced neuronal death. In addition, overexpression of nNOS in wild type CGN ameliorated alcohol-induced cell death. These results indicate that the neuroprotective effect of the cAMP pathway is mediated, in part, by the pathway's downstream target, the nNOS gene.
Collapse
Affiliation(s)
- Bahri Karaçay
- Department of Pediatrics Anatomy, University of Iowa College of Medicine Iowa City, Iowa 52242
| | - Guiying Li
- Department of Cell Biology, University of Iowa College of Medicine Iowa City, Iowa 52242
| | - Nicholas J. Pantazis
- Department of Cell Biology, University of Iowa College of Medicine Iowa City, Iowa 52242
- Department of Neuroscience Program, University of Iowa College of Medicine Iowa City, Iowa 52242
| | - Daniel J. Bonthius
- Department of Pediatrics Anatomy, University of Iowa College of Medicine Iowa City, Iowa 52242
- Department of Cell Biology, University of Iowa College of Medicine Iowa City, Iowa 52242
- Department of Neuroscience Program, University of Iowa College of Medicine Iowa City, Iowa 52242
- Department of Neurology, University of Iowa College of Medicine Iowa City, Iowa 52242
| |
Collapse
|
246
|
Mehta SL, Manhas N, Raghubir R. Molecular targets in cerebral ischemia for developing novel therapeutics. ACTA ACUST UNITED AC 2007; 54:34-66. [PMID: 17222914 DOI: 10.1016/j.brainresrev.2006.11.003] [Citation(s) in RCA: 539] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 11/09/2006] [Accepted: 11/10/2006] [Indexed: 11/20/2022]
Abstract
Cerebral ischemia (stroke) triggers a complex series of biochemical and molecular mechanisms that impairs the neurologic functions through breakdown of cellular integrity mediated by excitotoxic glutamatergic signalling, ionic imbalance, free-radical reactions, etc. These intricate processes lead to activation of signalling mechanisms involving calcium/calmodulin-dependent kinases (CaMKs) and mitogen-activated protein kinases (MAPKs) such as extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK). The distribution of these transducers bring them in contact with appropriate molecular targets leading to altered gene expression, e.g. ERK and JNK mediated early gene induction, responsible for activation of cell survival/damaging mechanisms. Moreover, inflammatory reactions initiated at the neurovascular interface and alterations in the dynamic communication between the endothelial cells, astrocytes and neurons are thought to substantially contribute to the pathogenesis of the disease. The damaging mechanisms may proceed through rapid nonspecific cell lysis (necrosis) or by active form of cell demise (apoptosis or necroptosis), depending upon the severity and duration of the ischemic insult. A systematic understanding of these molecular mechanisms with prospect of modulating the chain of events leading to cellular survival/damage may help to generate the potential strategies for neuroprotection. This review briefly covers the current status on the molecular mechanisms of stroke pathophysiology with an endeavour to identify potential molecular targets such as targeting postsynaptic density-95 (PSD-95)/N-methyl-d-aspartate (NMDA) receptor interaction, certain key proteins involved in oxidative stress, CaMKs and MAPKs (ERK, p38 and JNK) signalling, inflammation (cytokines, adhesion molecules, etc.) and cell death pathways (caspases, Bcl-2 family proteins, poly (ADP-ribose) polymerase-1 (PARP-1), apoptosis-inducing factor (AIF), inhibitors of apoptosis proteins (IAPs), heat shock protein 70 (HSP70), receptor interacting protein (RIP), etc., besides targeting directly the genes itself. However, selecting promising targets from various signalling cascades, for drug discovery and development is very challenging, nevertheless such novel approaches may lead to the emergence of new avenues for therapeutic intervention in cerebral ischemia.
Collapse
Affiliation(s)
- Suresh L Mehta
- Division of Pharmacology, Central Drug Research Institute, Chatter Manzil Palace, POB-173, Lucknow-226001, India
| | | | | |
Collapse
|
247
|
Mioduszewska B, Jaworski J, Szklarczyk AW, Klejman A, Kaczmarek L. Inducible cAMP early repressor (ICER)-evoked delayed neuronal death in the organotypic hippocampal culture. J Neurosci Res 2007; 86:61-70. [PMID: 17722060 DOI: 10.1002/jnr.21469] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Programmed cell death involving gene regulation and de novo protein synthesis is a major component of both normal development and a number of disease conditions. Hence, knowledge of its mechanisms, especially transcription factors, that regulate expression of the genes involved in neurodegenerative disorders is of great importance. cAMP-responsive element-binding protein (CREB) has repeatedly been implicated in the neuronal survival. In the present study we showed that inducible cAMP early repressor (ICER), an endogenous CREB antagonist, is expressed during both excitotoxic and spontaneous neuronal cell death in organotypic hippocampal slice cultures in vitro. Furthermore, overexpression of ICER via an adenoviral vector evoked neuronal cell loss in such cultures. The time course of ICER-dependent cell death was hippocampal subdivision specific, with dentate gyrus neurons dying mostly 3-7 days after the adenovector infection, followed by CA3, where neuronal death peaked after 7 days, and then CA1, where most neuronal death occurred after 7-14 days. These results underscore the usefulness of the organotypic cultures for studies of neurodegeneration and point to neuronal loss having a multifaceted nature in a complex cellular environment.
Collapse
|
248
|
Ryan MM, Lockstone HE, Huffaker SJ, Wayland MT, Webster MJ, Bahn S. Gene expression analysis of bipolar disorder reveals downregulation of the ubiquitin cycle and alterations in synaptic genes. Mol Psychiatry 2006; 11:965-78. [PMID: 16894394 DOI: 10.1038/sj.mp.4001875] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bipolar affective disorder is a severe psychiatric disorder with a strong genetic component but unknown pathophysiology. We used microarray technology to determine the expression of approximately 22,000 mRNA transcripts in post-mortem tissue from two brain regions in patients with bipolar disorder and matched healthy controls. Dorsolateral prefrontal cortex tissue from a cohort of 70 subjects and orbitofrontal cortex tissue from a separate cohort of 30 subjects was investigated. The final analysis included 30 bipolar and 31 control subjects for the dorsolateral prefrontal cortex and 10 bipolar and 11 control subjects for the orbitofrontal cortex. Differences between disease and control groups were identified using a rigorous statistical analysis with correction for confounding variables and multiple testing. In the orbitofrontal cortex, 393 differentially expressed transcripts were identified by microarray analysis and a representative subset was validated by quantitative real-time PCR. Pathway analysis revealed significant upregulation of genes involved in G-protein coupled receptor signalling and response to stimulus (in particular the immune response), while genes relating to the ubiquitin cycle and intracellular transport showed coordinated downregulation in bipolar disorder. Additionally, several genes involved in synaptic function were significantly downregulated in bipolar disorder. No significant changes in gene expression were observed in the dorsolateral prefrontal cortex using microarray analysis or quantitative real-time PCR. Our findings implicate the orbitofrontal cortex as a region prominently involved in bipolar disorder and indicate that diverse processes are affected. Overall, our results suggest that dysregulation of the ubiquitin pathway and synaptic function may be central to the disease process.
Collapse
Affiliation(s)
- M M Ryan
- Cambridge Centre for Neuropsychiatric Research, Institute of Biotechnology, University of Cambridge, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
249
|
Gruol DL, Quina LA, Netzeband JG, Nguyen D, Gullette CE. Developmental changes in Ca2+-regulated functions of early postnatal Purkinje neurons. J Neurosci Res 2006; 83:1381-92. [PMID: 16555300 DOI: 10.1002/jnr.20844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Ca(2+) influx through L-type Ca(2+) channels regulates several different cellular processes in developing Purkinje neurons, including activation of transcription factors and expression of cellular proteins. In the current studies, we examined the age dependence of these actions of Ca(2+) during the early developmental period. Purkinje neurons acutely isolated from postnatal day 4-8 rat pups were studied. We also examined the sensitivity of the Ca(2+)-regulated processes to a toxic environmental factor (ethanol) known to show age-dependent actions on developing Purkinje neurons. Results show that Ca(2+) activation of the transcription factor cAMP-responsive element binding protein (CREB) and Ca(2+)-induced alterations in the level of the apoptotic enzyme caspase 3 show both dose and age dependence in the early-developing Purkinje neurons. Interestingly, the age dependence was opposite for the two proteins. Ca(2+) regulation of calbindin, a Ca(2+) binding protein, was dose dependent but showed little age dependence. Exposure to ethanol altered Ca(2+) activation of pCREB in an age-dependent manner but did not alter Ca(2+) regulation of caspase 3 or calbindin levels. Taken together, these results show that the downstream effects of Ca(2+) signaling have age-dependent components during early Purkinje neuron development. This age dependence may play an important role in the normal developmental program and could contribute to the critical window of sensitivity observed for certain toxic agents during early development.
Collapse
Affiliation(s)
- D L Gruol
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California, USA.
| | | | | | | | | |
Collapse
|
250
|
Lee TH, Yang JT, Kato H, Wu JH. Hypertension downregulates the expression of brain-derived neurotrophic factor in the ischemia-vulnerable hippocampal CA1 and cortical areas after carotid artery occlusion. Brain Res 2006; 1116:31-8. [PMID: 16962081 DOI: 10.1016/j.brainres.2006.07.117] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Revised: 07/17/2006] [Accepted: 07/29/2006] [Indexed: 12/23/2022]
Abstract
We studied the effect of hypertension on brain damage and brain-derived neurotrophic factor (BDNF) expression in the hippocampal formation and cerebral cortex after permanent occlusion of bilateral common carotid arteries (CCA). Two groups of rats were used, including normotensive Wistar-Kyoto (WKY) rat and spontaneous hypertensive rat (SHR). Each group contained sham operation, 1 week and 4 weeks after bilateral CCA occlusion (n=5-10 in each time point). The blood pressure showed a significant elevation in WKY rats from 1 h after operation to 4 weeks before sacrifice (P<0.05), but was not changed in SHR (P>0.05). However, rectal temperature showed no significant change after operation in WKY rat and SHR (P>0.05) and showed no significant difference at any time point between WKY rat and SHR (P>0.05). Hematoxylin and eosin staining showed SHR had a significantly larger necrotic volume than WKY rats (n=10 in each group, 6044+/-6895 microm(3) vs. 144+/-174 microm(3), P<0.05) at 4 weeks after ischemia. In SHR, BDNF immunoreactivity and mRNA decreased significantly from 1 week to 4 weeks in both the hippocampal CA1 and cortical areas (P<0.01) but decreased transiently in dentate gyrus. However, in WKY rats, BDNF immunoreactivity and mRNA decreased transiently at 1 week (P<0.05) and recovered at 4 weeks after cerebral ischemia. Our study demonstrates that after bilateral CCA occlusion, preexisting hypertension may aggravate the brain injury and downregulate the expression of BDNF immunoreactivity and mRNA in the ischemia-vulnerable hippocampal CA1 and cortical areas but not in ischemia-resistant dentate gyrus.
Collapse
Affiliation(s)
- Tsong-Hai Lee
- Stroke Section, Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, College of Medicine, Chang Gung University, 5 Fu-Hsing St., Kuei-Shan, Tao-Yuan, 33333 Taiwan.
| | | | | | | |
Collapse
|