Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-27358653

ABSTRACT

BACKGROUND: Prenatal alcohol exposure is the leading preventable cause of behavioral and cognitive deficits, which may affect between 2 and 5 % of children in North America. While the underlying mechanisms of alcohol's effects on development remain relatively unknown, emerging evidence implicates epigenetic mechanisms in mediating the range of symptoms observed in children with fetal alcohol spectrum disorder (FASD). Thus, we investigated the effects of prenatal alcohol exposure on genome-wide DNA methylation in the NeuroDevNet FASD cohort, the largest cohort of human FASD samples to date. METHODS: Genome-wide DNA methylation patterns of buccal epithelial cells (BECs) were analyzed using the Illumina HumanMethylation450 array in a Canadian cohort of 206 children (110 FASD and 96 controls). Genotyping was performed in parallel using the Infinium HumanOmni2.5-Quad v1.0 BeadChip. RESULTS: After correcting for the effects of genetic background, we found 658 significantly differentially methylated sites between FASD cases and controls, with 41 displaying differences in percent methylation change >5 %. Furthermore, 101 differentially methylated regions containing two or more CpGs were also identified, overlapping with 95 different genes. The majority of differentially methylated genes were highly expressed at the level of mRNA in brain samples from the Allen Brain Atlas, and independent DNA methylation data from cortical brain samples showed high correlations with BEC DNA methylation patterns. Finally, overrepresentation analysis of genes with up-methylated CpGs revealed a significant enrichment for neurodevelopmental processes and diseases, such as anxiety, epilepsy, and autism spectrum disorders. CONCLUSIONS: These findings suggested that prenatal alcohol exposure is associated with distinct DNA methylation patterns in children and adolescents, raising the possibility of an epigenetic biomarker of FASD.

2.
Neurochem Int ; 96: 46-55, 2016 06.
Article in English | MEDLINE | ID: mdl-26939762

ABSTRACT

BACKGROUND: Significant protease activations have been reported after traumatic brain injury (TBI). These proteases are responsible for cleavage of transmembrane proteins in neurons, glial, and endothelial cells and this results in the release of their extracellular domains (ectodomains). METHODS: Two TBI models were employed here, representing both closed head injury (CHI) and open head injury (OHI). In situ zymography, immunohistochemistry, bright field and confocal microscopy, quantification of immunopositive cells and statistical analysis were applied. RESULTS: We found, using in situ zymography, that gelatinase activity of matrix metalloproteinases (MMP)-2 and MMP-9 was upregulated in cortex of both injury models. Using immunohistochemistry for several MPPs (Matrix metalloproteinases) and ADAMs (disintegrin and metalloproteinases), including MMP-2, -9, ADAM-10, -17, distinct patterns of induction were observed in the two TBI models. In closed head injury, an early increase in protein expression of MMP-2, -9 and ADAM-17 was found as early as 10 min post injury in cortex and peaked at 1 h for all 4 proteases examined. In contrast, after OHI the maximal expression was observed locally neighboring the impact site, at a later time-point, as long as 24 h after the injury for MMP-2 and MMP-9. Confocal microscopy revealed colocalization of the 4 proteases with the neuronal marker NeuN in CHI, but only MMP2 colocalized with NeuN in OHI. CONCLUSIONS: The findings may lead to a trauma-induced therapeutic strategy triggered soon after a primary insult to improve survival and to reduce brain damage following TBI.


Subject(s)
Craniocerebral Trauma/enzymology , Head Injuries, Closed/enzymology , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinase 9/physiology , ADAM17 Protein/physiology , Animals , Brain Injuries, Traumatic/enzymology , Brain Injuries, Traumatic/pathology , Craniocerebral Trauma/pathology , Head Injuries, Closed/pathology , Male , Mice , Mice, Inbred C57BL
3.
PLoS One ; 10(11): e0142296, 2015.
Article in English | MEDLINE | ID: mdl-26571498

ABSTRACT

TDP-43 (TAR DNA binding protein 43) is a heterogeneous nuclear ribonucleoprotein (hnRNP) that has been found to play an important role in neurodegenerative diseases. TDP-43's involvement in nuclear factor-kappaB pathways has been reported in both neurons and microglial cells. The NF-κB pathway targets hundreds of genes, many of which are involved in inflammation, immunity and cancer. p50/p65 (p50/RelA) heterodimers, as the major Rel complex in the NF-κB family, are induced by diverse external physiological stimuli and modulate transcriptional activity in almost all cell types. Both p65 and TDP-43 translocation occur through the classic nuclear transportation system. In this study, we report that TDP-43 overexpression prevents TNF-α induced p65 nuclear translocation in a dose dependent manner, and that this further inhibits p65 transactivation activity. The inhibition by TDP-43 does not occur through preventing IκB degradation but probably by competing for the nuclear transporter-importin α3 (KPNA4). This competition is dependent on the presence of the nuclear localization signal (NLS) in TDP-43. Silencing TDP-43 using a specific siRNA also increased p65 nuclear localization upon TNF-α stimulation, suggesting that endogenous TDP-43 may be a default suppressor of the NF-κB pathway. Our results indicate that TDP-43 may play an important role in regulating the levels of NF-κB activity by controlling the nuclear translocation of p65.


Subject(s)
DNA-Binding Proteins/physiology , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus , Animals , Binding, Competitive , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , I-kappa B Proteins/metabolism , MCF-7 Cells , Nuclear Localization Signals/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Protein Binding , Proteolysis , Transcriptional Activation , Tumor Necrosis Factor-alpha/physiology , alpha Karyopherins/metabolism
5.
BMC Med Genomics ; 6: 58, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24373378

ABSTRACT

BACKGROUND: The presence of an extra whole or part of chromosome 21 in people with Down syndrome (DS) is associated with multiple neurological changes, including pathological aging that often meets the criteria for Alzheimer's Disease (AD). In addition, trisomies have been shown to disrupt normal epigenetic marks across the genome, perhaps in response to changes in gene dosage. We hypothesized that trisomy 21 would result in global epigenetic changes across all participants, and that DS patients with cognitive impairment would show an additional epigenetic signature. METHODS: We therefore examined whole-genome DNA methylation in buccal epithelial cells of 10 adults with DS and 10 controls to determine whether patterns of DNA methylation were correlated with DS and/or cognitive impairment. In addition we examined DNA methylation at the APP gene itself, to see whether there were changes in DNA methylation in this population. Using the Illumina Infinium 450 K Human Methylation Array, we examined more than 485,000 CpG sites distributed across the genome in buccal epithelial cells. RESULTS: We found 3300 CpGs to be differentially methylated between the groups, including 495 CpGs that overlap with clusters of differentially methylated probes. In addition, we found 5 probes that were correlated with cognitive function including two probes in the TSC2 gene that has previously been associated with Alzheimer's disease pathology. We found no enrichment on chromosome 21 in either case, and targeted analysis of the APP gene revealed weak evidence for epigenetic impacts related to the AD phenotype. CONCLUSIONS: Overall, our results indicated that both Trisomy 21 and cognitive impairment were associated with distinct patterns of DNA methylation.


Subject(s)
Cognition , DNA Methylation , Down Syndrome/genetics , Down Syndrome/physiopathology , Adult , Amyloid beta-Protein Precursor/genetics , Case-Control Studies , Chromosomes, Human, Pair 21/genetics , CpG Islands/genetics , Female , Genomics , Humans , Male , Middle Aged , Principal Component Analysis
6.
Acta Neuropathol Commun ; 1: 49, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-24252504

ABSTRACT

BACKGROUND: TAR DNA-binding protein 43 (TDP-43) is a protein that is involved in the pathology of Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Lobar Degeneration (FTLD). In patients with these neurodegenerative diseases, TDP-43 does not remain in its normal nuclear location, but instead forms insoluble aggregates in both the nucleus and cytoplasm of affected neurons. RESULTS: We used high density peptide array analysis to identify regions in TDP-43 that are bound by TDP-43 itself and designed candidate peptides that might be able to reduce TDP-43 aggregation. We found that two of the synthetic peptides identified with this approach could effectively inhibit the formation of TDP-43 protein aggregates in a concentration-dependent manner in HeLa cells in which a mutated human TDP-43 gene was overexpressed. However, despite reducing aggregation, these peptides did not reduce or prevent cell death. Similar results were observed in HeLa cells treated with arsenite. Again we found reduced aggregation, in this case of wild type TDP-43, but no difference in cell death. CONCLUSIONS: Our results suggest that TDP-43 aggregation is associated with the cell death process rather than being a direct cause.


Subject(s)
Cell Death/physiology , DNA-Binding Proteins/metabolism , Arsenites/toxicity , Cell Death/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Cell Survival/physiology , DNA-Binding Proteins/genetics , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoblotting , In Situ Nick-End Labeling , Mutation , Protein Binding
7.
Brain Res ; 1530: 54-65, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23887054

ABSTRACT

Progranulin (PGRN) haploinsufficiency accounts for up to 10% of frontotemporal lobe dementia. PGRN has also been implicated in neuroinflammation in acute and chronic neurological disorders. Here we report that both protein and mRNA levels of cortical and hippocampal PGRN are significantly enhanced following pilocarpine-induced status epilepticus. We also identify intense PGRN immunoreactivity that colocalizes with CD11b in seizure-induced animals, suggesting that PGRN elevation occurs primarily in activated microglia and macrophages. To test the role of PGRN in activation of microglia/macrophages, we apply recombinant PGRN protein directly into the hippocampal formation, and observe no change in the number of CD11b(+) microglia/macrophages in the dentate gyrus. However, with pilocarpine-induced status epilepticus, PGRN application significantly increases the number of CD11b(+) microglia/macrophages in the dentate gyrus, without affecting the extent of hilar cell death. In addition, the number of CD11b(+) microglia/macrophages induced by status epilepticus is not significantly different between PGRN knockout mice and wildtype. Our findings suggest that status epilepticus induces PGRN expression, and that PGRN potentiates but is not required for seizure-induced microglia/macrophage activation.


Subject(s)
Hippocampus/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Microglia/metabolism , Status Epilepticus/metabolism , Animals , Cell Death/physiology , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Disease Models, Animal , Granulins , Male , Mice , Mice, Knockout , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Pilocarpine/pharmacology , Progranulins , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced
8.
Sci Signal ; 6(283): ra57, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23838184

ABSTRACT

Secretory trafficking through the Golgi complex is critical for neuronal development, function, and stress response. Altered secretion is associated with the pathogenesis of various neurological diseases. We found that c-Jun amino-terminal kinase 3 (JNK3) inhibited secretory trafficking by promoting the depletion of phosphatidylinositol 4-phosphate (PI4P) in the Golgi complex of COS7 cells and primary rat neurons. Exposure of cultured primary rat neurons to excitotoxic concentrations of NMDA (N-methyl-d-aspartate), an agonist of a class of ionotropic glutamate receptors, or overexpression of zD17 (a palmitoyl transferase) resulted in JNK3 palmitoylation and association with the Golgi complex. Analysis of mutant constructs of JNK3 indicated that Golgi association was independent of its kinase activity but depended on its palmitoylation. The association of JNK3 with the Golgi in cultured neurons decreased the secretory trafficking of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluR1 (glutamate receptor subunit 1), a component of ionotropic glutamate receptors found at glutamatergic synapses. Palmitoylated JNK3 bound to the phosphatase Sac1, increasing its abundance at the Golgi and thereby decreasing the abundance of PI4P, a lipid necessary for post-Golgi trafficking. Disrupting the JNK3-Sac1 interaction with two synthetic peptides prevented the loss of surface GluR1 and preserved synaptic integrity in cultured neurons exposed to NMDA. Together, our results suggest that JNK3 participates in an adaptive response to neuronal hyperexcitation by impeding secretory trafficking at the Golgi complex.


Subject(s)
Mitogen-Activated Protein Kinase 10/metabolism , Neurons/enzymology , Stress, Physiological/physiology , Synapses/metabolism , Acyltransferases/biosynthesis , Acyltransferases/genetics , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , COS Cells , Chlorocebus aethiops , Excitatory Amino Acid Agonists/pharmacology , Golgi Apparatus/genetics , Inositol Polyphosphate 5-Phosphatases , Lipoylation/drug effects , Lipoylation/physiology , Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Mitogen-Activated Protein Kinase 10/genetics , Mutation , N-Methylaspartate/pharmacology , Neurons/cytology , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Rats , Rats, Wistar , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Stress, Physiological/drug effects , Synapses/genetics
9.
J Neurosci ; 31(33): 11980-91, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849558

ABSTRACT

Although the palmitoyl acyltransferase (PAT) zinc-finger DHHC containing 17 (zD17) has been implicated in genetic neurological disorders by regulating protein palmitoylation, the role of zD17 in acute brain injury remains unknown. Here, we report that zD17 contributes to acute ischemic brain injury via a mechanism independent of its PAT activity. We have found that zD17 directly interacts with c-Jun N terminus kinase (JNK) to form a signaling module for JNK activation. Pathological stressors induce the zD17-JNK interaction, which promotes downstream neuronal cell death signals. We have developed novel peptides targeting the JNK-interacting motif on zD17 to selectively block the enhancement of the zD17-JNK interaction and the activation of JNK isoforms 2 and 3. Application of these peptides successfully blocks JNK activation and neuronal cell death pathways, protects cultured neurons from excitotoxicity, and dramatically reduces brain damage and behavioral deficits in a rat model of focal ischemic stroke. Our findings indicate zD17 as a key player in ischemic stroke and suggest the potential therapeutic value of targeting the zD17-JNK interaction for acute brain injury.


Subject(s)
Acyltransferases/physiology , Adaptor Proteins, Signal Transducing/physiology , Brain Injuries/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/physiology , Nerve Tissue Proteins/physiology , Neurons/enzymology , Amino Acid Motifs/physiology , Animals , Brain Injuries/prevention & control , Cells, Cultured , Enzyme Activation/physiology , HEK293 Cells , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Isoenzymes/physiology , JNK Mitogen-Activated Protein Kinases/chemistry , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar
10.
Brain Res ; 1366: 1-8, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20888804

ABSTRACT

Null mutations in the progranulin gene (PGRN) have been identified as a major cause of frontotemporal dementia with ubiquitinated inclusions. In this disorder, ubiquitinated, aggregated protein inclusions of a normally nuclear-located RNA processing protein called TAR DNA binding protein (TDP-43) accumulate in the neuronal cytoplasm (FTLD-TDP). To determine whether aspects of this clinical pathology can be established in primary cultures of mouse cortical neurons, PGRN levels were knocked down in neuronal cultures using lentiviral vectors to introduce mouse PGRN-siRNA constructs and subsequently rescued by overexpressing PGRN using a human PGRN-expressing lentiviral vector. The depletion of PGRN enhanced caspase-3 activation, and the PGRN-deficient neurons demonstrated enhanced vulnerability to normally sublethal doses of N-methyl-D-aspartic acid (NMDA) and hydrogen peroxide (H(2)O(2)). TDP-43 protein levels were markedly increased in the cytoplasm of PGRN-deficient neurons relative to nuclear levels, which is similar to observations in the brains of FTLD-TDP patients. Our results establish a neuronal culture model of the PGRN deficiency, which displays some of the important phenotypic characteristics of the early stages of the disease. The results further suggest that the seeds of this form of frontotemporal dementia may be sown early in life.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Intercellular Signaling Peptides and Proteins/deficiency , Neurons/metabolism , Animals , Caspase 3/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Excitatory Amino Acid Agonists/pharmacology , Female , Gene Expression Regulation/drug effects , Granulins , Humans , Hydrogen Peroxide/pharmacology , Indoles , Intercellular Signaling Peptides and Proteins/genetics , Mice , N-Methylaspartate/pharmacology , Neurons/drug effects , Oxidants/pharmacology , Pregnancy , Progranulins , Protein Transport/drug effects , Protein Transport/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tetrazolium Salts , Thiazoles , Transfection/methods
11.
Invest Ophthalmol Vis Sci ; 51(12): 6207-19, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20702827

ABSTRACT

PURPOSE: To investigate the morphologic and molecular consequences of 30- and 90-minute central retinal artery ligation (CRAL)-induced retinal ischemia, followed by 3 and 12 hours of reperfusion, and to identify potential targets for therapy. METHODS: Retinal ischemia was induced for 30 and 90 minutes by ligating the rat central retinal artery, and corresponding effects were examined histologically, immunocytochemically, and molecularly at 3 hours and 12 hours of reperfusion. Patterns of gene expression revealed significantly upregulated and downregulated genes by gene array analyses and were verified by quantitative RT-PCR. Functional pathways were correlated using gene set enrichment analysis. RESULTS: Substantial morphologic changes occurred from 3 hours to 7 days after CRAL in rats, resulting in a cellular loss in most retinal layers, particularly in inner nuclear and ganglion cell layers. After 30 minutes of CRAL and 3 hours of reperfusion, transcription-related genes such as ATF3, ID2, Klf4, BTG2, c-Fos, and c-Jun were activated. After 12 hours of reperfusion, the genes associated with kinase and caspase molecular pathways-including MAP kinases, Casp3 and Casp9-were upregulated. CRAL of 90 minutes and 3 hours of reperfusion induced glycolysis and gluconeogenesis-related genes such as G6PC. However, prolonged reperfusion of 12 hours was characterized by prominent activation of apoptosis-related genes, including Tp53, Akt1, Akt3, Pik3R1, Prkcb1, caspases (Casp3, Casp7, Casp9), and TNF. CONCLUSIONS: CRAL is a clinically relevant retinal ischemia model, and gene expression analysis can provide information regarding the molecular mechanisms underlying the pathophysiological processes during retinal ischemia. In addition, CRAL represents an effective experimental model with which to study retinal inflammation, development, aging, and, neurodegeneration.


Subject(s)
Eye Proteins/genetics , Gene Expression Regulation/physiology , Ischemia/genetics , Reperfusion Injury/genetics , Retina/metabolism , Retinal Artery/physiology , Animals , Apoptosis Regulatory Proteins/genetics , Gene Expression Profiling , In Situ Nick-End Labeling , Ischemia/metabolism , Ischemia/physiopathology , Kruppel-Like Factor 4 , Ligation , Male , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred Lew , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
12.
Nat Med ; 15(12): 1399-406, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19966780

ABSTRACT

Excitotoxic neuronal damage caused by overactivation of N-methyl-D-aspartate glutamate receptors (NMDARs) is thought to be a principal cause of neuronal loss after stroke and brain trauma. Here we report that activation of sterol regulatory element binding protein-1 (SREBP-1) transcription factor in affected neurons is an essential step in NMDAR-mediated excitotoxic neuronal death in both in vitro and in vivo models of stroke. The NMDAR-mediated activation of SREBP-1 is a result of increased insulin-induced gene-1 (Insig-1) degradation, which can be inhibited with an Insig-1-derived interference peptide (Indip) that we have developed. Using a focal ischemia model of stroke, we show that systemic administration of Indip not only prevents SREBP-1 activation but also substantially reduces neuronal damage and improves behavioral outcome. Our study suggests that agents that reduce SREBP-1 activation such as Indip may represent a new class of neuroprotective therapeutics against stroke.


Subject(s)
Brain Ischemia/physiopathology , Cerebral Cortex/blood supply , Receptors, N-Methyl-D-Aspartate/physiology , Sterol Regulatory Element Binding Protein 1/physiology , Animals , Cell Line , Cells, Cultured , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/physiology , Rats , Rats, Sprague-Dawley
13.
Eur J Neurosci ; 30(1): 35-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19508696

ABSTRACT

The AMPA receptor (AMPAR) subunits GluR1 and GluR2 show different properties in central neurons and affect AMPAR trafficking via distinct mechanisms. This subunit-specificity is partly achieved by recruiting unique protein modifications on different subunits. Here, we show that palmitoylation of GluR1 and GluR2 subunits also displays subunit-specific properties and functions. Our findings indicate that GluR1 palmitoylation requires dynamic anterograde transport from the endoplasmic reticulum (ER) to the Golgi apparatus. In contrast, GluR2 subunits are primarily palmitoylated locally in the ER as immature receptors, and an intact microtubule network is required for their palmitoylation. Interestingly, the majority of palmitoylated GluR2 subunits are not associated with GluR1 subunits. We found that preventing palmitoylation results in loss of mature GluR2, but leaves GluR1 intact, as palmitoylation on GluR2 in the ER prevents their sorting to the lysosome after receptor maturation. Moreover, palmitoylation on GluR1 and GluR2 subunits responds differently to neuronal activity. Blocking neuronal activity by tetrodotoxin increased the pool size of palmitoylated GluR2, but not GluR1. Acute stimulation by NMDA and AMPA also differentially affect AMPAR palmitoylation in a subunit-specific manner. The present findings thus indicate that AMPAR palmitoylation is a subunit-specific process that contributes to its regulation and trafficking.


Subject(s)
Biological Transport/physiology , Lipoylation/physiology , Neurons/physiology , Receptors, AMPA/metabolism , Animals , Cell Line, Transformed , Cells, Cultured , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Humans , Lysosomes/metabolism , Microtubules/metabolism , N-Methylaspartate/metabolism , Neurons/drug effects , Protein Stability , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, AMPA/genetics , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
14.
Brain Res ; 1250: 49-62, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19041854

ABSTRACT

We report the first demonstration that Krüpple-like factor 4 (KLF4) mRNA is dramatically and rapidly upregulated by NMDA application in primary cortical neuron cultures. We also report that NMDA induced significant and transient upregulation of KLF4 protein expression, in both cortical neuron cultures and native brain slices. The increase of KLF4 mRNA and protein expression in response to NMDA was time-dependent, and required NMDA receptor-mediated Ca(2+) influx. In addition, AMPA exposure caused a time-dependent increase in KLF4 mRNA expression, which was also Ca(2+)-dependent and involved activation of AMPA/kainate receptors and L-type voltage-sensitive calcium channels. To assess the downstream signaling pathways and functions of KLF4 activation, we used lentiviral vectors to induce ectopic overexpression of KLF4 in cultured neurons. KLF4 overexpression induced the activation of caspase-3 after a normally subtoxic dose of NMDA (10 microM). KLF4 overexpression also increased both protein and mRNA levels of the cell cycle protein cyclin D1, but reduced p21(Waf1/Cip1) protein levels. After the NMDA treatment, cyclin D1 levels increased after a short delay (4 h), but fell back to control levels after 20 h. The effects of NMDA and KLF4 overexpression on cyclin D1 induction were additive. We conclude that glutamatergic stimulation can trigger rapid elevation of KLF4 mRNA and protein levels, and that the overexpression of KLF4 can regulate neuronal cell cycle proteins and sensitize neurons to NMDA-induced caspase-3 activity.


Subject(s)
Caspase 3/metabolism , Glutamic Acid/physiology , Kruppel-Like Transcription Factors/metabolism , N-Methylaspartate/pharmacology , Neurons/physiology , Animals , Blotting, Western , Cells, Cultured , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Enzyme Activation , Hippocampus/metabolism , Immunohistochemistry , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation
15.
Brain Res ; 1085(1): 68-76, 2006 Apr 26.
Article in English | MEDLINE | ID: mdl-16581040

ABSTRACT

Anisomycin is both a well-established protein synthesis inhibitor and a potent activator of the p38/JNK MAPK pathway. It has been used to block the late phase of long-term potentiation (LTP) and long-term depression (LTD) in hippocampus. In this study, we have found that anisomycin produces a time-dependent decline in the magnitude of the field EPSP (fEPSP) in acute brain slices of mouse primary visual cortex. This anisomycin-mediated fEPSP depression occludes NMDA receptor-dependent LTD induced by low-frequency stimulation (LFS). In contrast, two other protein synthesis inhibitors, emetine and cycloheximide, have no effect either on baseline synaptic transmission or on LTD. Moreover, the decline of the fEPSP caused by anisomycin can be rescued by the application of the p38 inhibitor SB203580 but not by the JNK inhibitor SP600125. These results indicate that activation of p38 MAPK by anisomycin induces LTD and subsequently occludes electrically induced LTD. Also, the occlusion of LFS-LTD by anisomycin suggests that common mechanisms may be shared between the two forms of synaptic depression. Consistent with this view, bath application of a membrane permeant peptide derived from the carboxyl tail of GluR2 subunit of AMPA receptor, which specifically blocks regulated AMPA receptor endocytosis, thereby preventing the expression of LFS-induced LTD, significantly reduced the anisomycin-induced decline of the fEPSP. In conclusion, our results indicate that anisomycin produces long-lasting depression of AMPA receptor-mediated synaptic transmission by activating p38 MAPK-mediated endocytosis of APMA receptors in mouse primary visual cortex.


Subject(s)
Anisomycin/pharmacology , Long-Term Synaptic Depression/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Visual Cortex/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Blotting, Western/methods , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Mice , Mice, Inbred C57BL , Synaptic Transmission/drug effects
16.
Free Radic Biol Med ; 40(6): 960-7, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16540391

ABSTRACT

Oxidative stress plays an important role in the pathogenesis of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). Bilirubin is regarded today as a potent antioxidant. Recent studies show that the potent antioxidant actions of bilirubin reflect an amplification mechanism whereby biliverdin reductase (BVR) physiologically regenerates bilirubin in a catalytic cycle. We hypothesized that BVR might prove to be a new effective target for the treatment of free radical-mediated diseases. In this study, we demonstrated that treatment with BVR ameliorated both clinical and pathological signs of EAE more efficiently than treatments with traditional antioxidant enzymes. In vitro, interference with cellular BVR activity by siRNA elicited greater increases in reactive oxygen species and cell death than interference with the activities of other antioxidant enzymes. Further studies showed that BVR surpasses other enzymes by the multifactorial functions of its only end product, bilirubin, including anti-complement activity, and an activity that inhibits antibody-dependent cell-mediated cytotoxicity of lymphocytes. Since BVR regenerates bilirubin in a redox cycle without significantly increasing the concentration of bilirubin, our results suggest that BVR may represent a novel strategy for the treatment of multiple sclerosis and other oxidative stress-mediated diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Oxidoreductases Acting on CH-CH Group Donors/physiology , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antioxidants/metabolism , Bilirubin/pharmacology , Cytoprotection/physiology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Enzyme Induction , Glutathione/pharmacology , Hemolysis/drug effects , Humans , Male , Myelitis/pathology , Myelitis/prevention & control , Neuroblastoma , Oxidative Stress/drug effects , Rats , Rats, Inbred Lew , Tumor Cells, Cultured
17.
Cereb Cortex ; 13(10): 1094-109, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12967926

ABSTRACT

Zinc is packaged in, and released from, a subset of glutamatergic synapses in the mammalian telencephalon where it has been shown to act as a potent neuromodulator. In order to establish the functional role for zincergic neurons in visual cortical function and plasticity we have compared the topographic distribution of zincergic terminals in the primary visual cortex (V1) of normal adult vervet monkeys (Cercopithicus aethiops) to that in monkeys monocularly deprived of visual input for short (24 h) or long (3 months) survival times. In normal animals, staining levels for zinc were highest in layers 1-3, 4b, 5 and 6 and lowest in layers 4a and 4c. The laminar and tangential patterns of zinc staining were complementary to staining patterns demonstrated using cytochrome oxidase (CO) histochemistry. Following 3 months of monocular deprivation by enucleation, levels of zinc staining in layers 3, 4calpha and 6a were heterogeneously reduced, clearly revealing the ocular dominance pattern in V1. When compared with the pattern of CO staining, levels of both CO and zinc were reduced in cortical territory innervated by the enucleated eye. Zinc histochemistry also revealed the ocular dominance pattern after only 24 h of monocular impulse blockade induced by enucleation or intravitreal tetrodotoxin infusion. However, by either means of deprivation for 24 h, levels of zinc were increased in deprived-eye stripes relative to nondeprived-eye stripes. These results indicate that zincergic terminals demarcate distinct compartments in the primate visual cortex. Furthermore, levels of synaptic zinc are rapidly and dynamically regulated, suggesting that zinc and/or zincergic neurons participate in mediating activity-dependent changes in the organization of the adult neocortex.


Subject(s)
Neurons/metabolism , Presynaptic Terminals/metabolism , Sensory Deprivation/physiology , Visual Cortex/metabolism , Zinc/metabolism , Animals , Chlorocebus aethiops , Eye Enucleation/methods , Male , Neurons/chemistry , Presynaptic Terminals/chemistry , Visual Cortex/chemistry , Zinc/analysis
18.
J Neuroimmunol ; 139(1-2): 27-35, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12799017

ABSTRACT

Increasing evidence shows that oxidative stress plays an important role in the pathogenesis of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). In recent years, bilirubin has been demonstrated to be a potent antioxidant in vitro. In this study, we administered bilirubin to rats with acute and chronic EAE. Bilirubin prevented both acute and chronic EAE effectively. More significantly, bilirubin suppressed ongoing clinical EAE and halted EAE progression when given after disease onset. Subsequent histological examination showed that if administered to rats before the onset of EAE, bilirubin interfered with the invasion of inflammatory cells into the central nervous system (CNS) because it protected the blood-brain barrier (BBB) from free radical-induced permeability changes. However, in some cases, inflammation still occurred even when no clinical illness was observed. In rats with treatment initiated after the onset of EAE, despite the clinical improvements, treatment with bilirubin did not reduce the degree of CNS inflammation, or change cytokine expression in CNS lesions, indicating a lack of immunosuppressive effect of this treatment. By contrast, bilirubin treatment significantly alleviated oxidative damage in the spinal cord, and the clinical signs of EAE correlated well with the degree of oxidative injury in the lesions. Our results suggest that free radicals play an important role in the final effector stages of EAE, and that antioxidant therapies may have potential for the treatment of MS.


Subject(s)
Antioxidants/pharmacology , Bilirubin/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/drug therapy , Oxidative Stress/drug effects , Animals , Antioxidants/therapeutic use , Bilirubin/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/metabolism , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Cytokines/drug effects , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Drug Administration Schedule , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Free Radicals/antagonists & inhibitors , Free Radicals/metabolism , Male , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/immunology , Rats , Rats, Inbred Lew , Reaction Time/drug effects , Reaction Time/immunology , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/metabolism , Treatment Outcome
19.
Am J Pathol ; 162(5): 1639-50, 2003 May.
Article in English | MEDLINE | ID: mdl-12707048

ABSTRACT

Evidence has shown that excitotoxicity may contribute to the loss of central nervous system axons and oligodendrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Because dendrites and synapses are vulnerable to excitotoxicity, we examined these structures in acute and chronic models of EAE. Immunostaining for microtubule-associated protein-2 showed that extensive dendritic beading occurred in the white matter of the lumbosacral spinal cord (LSSC) during acute EAE episodes and EAE relapses. Retrograde labeling confirmed that most motoneuron dendrites were beaded in the white matter of the LSSC in acute EAE. In contrast, only mild swelling was observed in the gray matter of the LSSC. Dendritic beading showed marked recovery during EAE remission and after EAE recovery. In addition, synaptophysin, synapsin I, and PSD-95 immunoreactivities were significantly reduced in both the gray and white matter of the LSSC during acute EAE episodes and EAE relapses, but showed partial recovery during EAE remission and after EAE recovery. Pathologically, both dendritic beading and the reduction in synaptic protein immunoreactivity were well correlated with inflammatory cell infiltration in the LSSC at different EAE stages. We propose that dendritic and synaptic damage in the spinal cord may contribute to the neurological deficits in EAE.


Subject(s)
Dendrites/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Spinal Cord/pathology , Synapses/pathology , Acute Disease , Animals , Axonal Transport , Chronic Disease , Disease Models, Animal , Female , Guinea Pigs , Male , Motor Neurons/pathology , Rats , Rats, Inbred Lew
20.
J Immunol ; 169(3): 1561-9, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12133985

ABSTRACT

Fas ligand (FasL) is an essential molecule strongly expressed in some immunoprivileged sites, but is expressed at very low levels in normal CNS. In this study, acute experimental autoimmune encephalomyelitis (EAE) was induced in Lewis rats with guinea pig myelin basic protein. Intrathecal infusion of recombinant FasL before EAE onset dose dependently suppressed acute EAE and alleviated pathological inflammation in lumbosacral spinal cord. This treatment greatly increased apoptosis in CNS inflammatory cells, but did not inhibit systemic immune response to myelin basic protein. Systemic administration of a similar dose of rFasL was ineffective. In vitro, encephalitogenic T cells were highly sensitive to rFasL-induced cell death, and activated macrophages were also susceptible. In addition, in vitro rFasL treatment potentiated the immunosuppressive property of rat cerebrospinal fluid. We conclude that intrathecal infusion of rFasL eliminated the initial wave of infiltrating T cells and macrophages, and therefore blocked the later recruitment of inflammatory cells into CNS. Although Fas receptor expression was observed on spinal cord neurons, astrocytes, and oligodendrocytes, no damage to these cells or to the myelin structure was detected after rFasL infusion.


Subject(s)
Brain/drug effects , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immunosuppressive Agents/pharmacology , Membrane Glycoproteins/pharmacology , Animals , Apoptosis , Brain/immunology , Cell Line , Fas Ligand Protein , In Situ Nick-End Labeling , Injections, Spinal , Macrophage Activation/drug effects , Male , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/therapeutic use , Myelin Sheath/drug effects , Rats , Rats, Inbred Lew , Recombinant Proteins/pharmacology , Spinal Cord/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...