Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Front Cell Dev Biol ; 12: 1371568, 2024.
Article En | MEDLINE | ID: mdl-38606319

The mammalian brain, especially the cerebral cortex, has evolved to increase in size and complexity. The proper development of the cerebral cortex requires the coordination of several events, such as differentiation and migration, that are essential for forming a precise six-layered structure. We have previously reported that Cdk5-mediated phosphorylation of JIP1 at T205 modulates axonal out-growth. However, the spatiotemporal expression patterns and functions of these three genes (Cdk5, Cdk5r1 or p35, and Mapk8ip1 or JIP1) in distinct cell types during cortical development remain unclear. In this study, we analyzed single-cell RNA-sequencing data of mouse embryonic cortex and discovered that Cdk5, p35, and JIP1 are dynamically expressed in intermediate progenitors (IPs). Pseudotime analysis revealed that the expression of these three genes was concomitantly upregulated in IPs during neuronal migration and differentiation. By manipulating the expression of JIP1 and phospho-mimetic JIP1 using in utero electroporation, we showed that phosphorylated JIP1 at T205 affected the temporal migration of neurons.

2.
BMC Biol ; 20(1): 115, 2022 05 17.
Article En | MEDLINE | ID: mdl-35581583

BACKGROUND: Activated Cdk5 regulates a number of processes during nervous system formation, including neuronal differentiation, growth cone stabilization, and axonal growth. Cdk5 phosphorylates its downstream substrates located in axonal growth cones, where the highly expressed c-Jun N-terminal kinase (JNK)-interacting protein1 (JIP1) has been implicated as another important regulator of axonal growth. In addition, stringent control of the level of intracellular domain of Notch1 (Notch1-IC) plays a regulatory role in axonal outgrowth during neuronal differentiation. However, whether Cdk5-JIP1-Notch1 cooperate to regulate axonal outgrowth, and the mechanism of such joint contribution to this pathway, is presently unknown, and here we explore their potential interaction. RESULTS: Our interactome screen identified JIP1 as an interactor of p35, a Cdk5 activator, and we sought to explore the relationship between Cdk5 and JIP1 on the regulation of axonal outgrowth. We demonstrate that JIP1 phosphorylated by Cdk5 at Thr205 enhances axonal outgrowth and a phosphomimic JIP1 rescues the axonal outgrowth defects in JIP1-/- and p35-/- neurons. Axonal outgrowth defects caused by the specific increase of Notch1 in JIP1-/- neurons are rescued by Numb-mediated inhibition of Notch1. Finally, we demonstrate that Cdk5 phosphorylation of JIP1 further amplifies the phosphorylation status of yet another Cdk5 substrate E3-ubiquitin ligase Itch, resulting in increased Notch1 ubiquitination. CONCLUSIONS: Our findings identify a potentially critical signaling axis involving Cdk5-JIP1-Itch-Notch1, which plays an important role in the regulation of CNS development. Future investigation into the way this pathway integrates with additional pathways regulating axonal growth will further our knowledge of normal central nervous system development and pathological conditions.


Neurons , Signal Transduction , Cells, Cultured , Neurons/metabolism , Phosphorylation , Signal Transduction/physiology
3.
Acta Neuropathol ; 141(5): 725-754, 2021 05.
Article En | MEDLINE | ID: mdl-33694021

The mechanisms by which parkin protects the adult human brain from Parkinson disease remain incompletely understood. We hypothesized that parkin cysteines participate in redox reactions and that these are reflected in its posttranslational modifications. We found that in post mortem human brain, including in the Substantia nigra, parkin is largely insoluble after age 40 years; this transition is linked to its oxidation, such as at residues Cys95 and Cys253. In mice, oxidative stress induces posttranslational modifications of parkin cysteines that lower its solubility in vivo. Similarly, oxidation of recombinant parkin by hydrogen peroxide (H2O2) promotes its insolubility and aggregate formation, and in exchange leads to the reduction of H2O2. This thiol-based redox activity is diminished by parkin point mutants, e.g., p.C431F and p.G328E. In prkn-null mice, H2O2 levels are increased under oxidative stress conditions, such as acutely by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxin exposure or chronically due to a second, genetic hit; H2O2 levels are also significantly increased in parkin-deficient human brain. In dopamine toxicity studies, wild-type parkin, but not disease-linked mutants, protects human dopaminergic cells, in part through lowering H2O2. Parkin also neutralizes reactive, electrophilic dopamine metabolites via adduct formation, which occurs foremost at the primate-specific residue Cys95. Further, wild-type but not p.C95A-mutant parkin augments melanin formation in vitro. By probing sections of adult, human midbrain from control individuals with epitope-mapped, monoclonal antibodies, we found specific and robust parkin reactivity that co-localizes with neuromelanin pigment, frequently within LAMP-3/CD63+ lysosomes. We conclude that oxidative modifications of parkin cysteines are associated with protective outcomes, which include the reduction of H2O2, conjugation of reactive dopamine metabolites, sequestration of radicals within insoluble aggregates, and increased melanin formation. The loss of these complementary redox effects may augment oxidative stress during ageing in dopamine-producing cells of mutant PRKN allele carriers, thereby enhancing the risk of Parkinson's-linked neurodegeneration.


Aging/metabolism , Dopamine/metabolism , Mesencephalon/metabolism , Nerve Degeneration/metabolism , Ubiquitin-Protein Ligases/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Aging/pathology , Animals , Child , Child, Preschool , Female , Humans , Male , Mesencephalon/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Degeneration/pathology , Oxidation-Reduction , Young Adult
4.
Exp Neurobiol ; 29(1): 70-79, 2020 Feb 29.
Article En | MEDLINE | ID: mdl-32122109

Many previous studies have shown reduced glucose uptake in the ischemic brain. In contrast, in a permanent unilateral common carotid artery occlusion (UCCAO) mouse model, our pilot experiments using 18F-fluorodeoxyglucose positron emission tomography (FDG PET) revealed that a subset of mice exhibited conspicuously high uptake of glucose in the ipsilateral hemisphere at 1 week post-occlusion (asymmetric group), whereas other mice showed symmetric uptake in both hemispheres (symmetric group). Thus, we aimed to understand the discrepancy between the two groups. Cerebral blood flow and histological/metabolic changes were analyzed using laser Doppler flowmetry and immunohistochemistry/Western blotting, respectively. Contrary to the increased glucose uptake observed in the ischemic cerebral hemisphere on FDG PET (p<0.001), cerebral blood flow tended to be lower in the asymmetric group than in the symmetric group (right to left ratio [%], 36.4±21.8 vs. 58.0±24.8, p=0.059). Neuronal death was observed only in the ischemic hemisphere of the asymmetric group. In contrast, astrocytes were more activated in the asymmetric group than in the symmetric group (p<0.05). Glucose transporter-1, and monocarboxylate transporter-1 were also upregulated in the asymmetric group, compared with the symmetric group (p<0.05, respectively). These results suggest that the increased FDG uptake was associated with relatively severe ischemia, and glucose transporter-1 upregulation and astrocyte activation. Glucose metabolism may thus be a compensatory mechanism in the moderately severe ischemic brain.

5.
J Biol Chem ; 294(21): 8617-8629, 2019 05 24.
Article En | MEDLINE | ID: mdl-30967472

We previously reported that the cell cycle-related cyclin-dependent kinase 4-retinoblastoma (RB) transcriptional corepressor pathway is essential for stroke-induced cell death both in vitro and in vivo However, how this signaling pathway induces cell death is unclear. Previously, we found that the cyclin-dependent kinase 4 pathway activates the pro-apoptotic transcriptional co-regulator Cited2 in vitro after DNA damage. In the present study, we report that Cited2 protein expression is also dramatically increased following stroke/ischemic insult. Critically, utilizing conditional knockout mice, we show that Cited2 is required for neuronal cell death, both in culture and in mice after ischemic insult. Importantly, determining the mechanism by which Cited2 levels are regulated, we found that E2F transcription factor (E2F) family members participate in Cited2 regulation. First, E2F1 expression induced Cited2 transcription, and E2F1 deficiency reduced Cited2 expression. Moreover, determining the potential E2F-binding regions on the Cited2 gene regulatory sequence by ChIP analysis, we provide evidence that E2F1/4 proteins bind to this DNA region. A luciferase reporter assay to probe the functional outcomes of this interaction revealed that E2F1 activates and E2F4 inhibits Cited2 transcription. Moreover, we identified the functional binding motif for E2F1 in the Cited2 gene promoter by demonstrating that mutation of this site dramatically reduces E2F1-mediated Cited2 transcription. Finally, E2F1 and E2F4 regulated Cited2 expression in neurons after stroke-related insults. Taken together, these results indicate that the E2F-Cited2 regulatory pathway is critically involved in stroke injury.


E2F1 Transcription Factor/metabolism , E2F4 Transcription Factor/metabolism , Gene Expression Regulation , Neurons/metabolism , Repressor Proteins/biosynthesis , Stroke/metabolism , Trans-Activators/biosynthesis , Amino Acid Motifs , Animals , Cell Death , E2F1 Transcription Factor/genetics , E2F4 Transcription Factor/genetics , Mice , Mice, Transgenic , Neurons/pathology , Repressor Proteins/genetics , Stroke/genetics , Stroke/pathology , Trans-Activators/genetics
6.
Science ; 357(6354): 891-898, 2017 09 01.
Article En | MEDLINE | ID: mdl-28860381

Copy number mutations implicate excess production of α-synuclein as a possibly causative factor in Parkinson's disease (PD). Using an unbiased screen targeting endogenous gene expression, we discovered that the ß2-adrenoreceptor (ß2AR) is a regulator of the α-synuclein gene (SNCA). ß2AR ligands modulate SNCA transcription through histone 3 lysine 27 acetylation of its promoter and enhancers. Over 11 years of follow-up in 4 million Norwegians, the ß2AR agonist salbutamol, a brain-penetrant asthma medication, was associated with reduced risk of developing PD (rate ratio, 0.66; 95% confidence interval, 0.58 to 0.76). Conversely, a ß2AR antagonist correlated with increased risk. ß2AR activation protected model mice and patient-derived cells. Thus, ß2AR is linked to transcription of α-synuclein and risk of PD in a ligand-specific fashion and constitutes a potential target for therapies.


Gene Expression Regulation , Parkinson Disease/ethnology , Parkinson Disease/genetics , Receptors, Adrenergic, beta-2/metabolism , alpha-Synuclein/genetics , Acetylation , Adrenergic beta-1 Receptor Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Albuterol/pharmacology , Albuterol/therapeutic use , Animals , Cell Line, Tumor , Enhancer Elements, Genetic , Gene Expression Regulation/drug effects , Histones/metabolism , Humans , Ligands , Mice , Neuroprotective Agents/pharmacology , Norway/ethnology , Parkinson Disease/drug therapy , Promoter Regions, Genetic , Propranolol/pharmacology , Propranolol/therapeutic use , Receptors, Adrenergic, beta-2/genetics , Risk , Substantia Nigra/metabolism , Transcription, Genetic/drug effects
7.
J Neurosci ; 37(28): 6729-6740, 2017 07 12.
Article En | MEDLINE | ID: mdl-28607169

Dysregulation of cell cycle machinery is implicated in a number of neuronal death contexts, including stroke. Increasing evidence suggests that cyclin-dependent kinases (Cdks) are inappropriately activated in mature neurons under ischemic stress conditions. We previously demonstrated a functional role for the cyclin D1/Cdk4/pRb (retinoblastoma tumor suppressor protein) pathway in delayed neuronal death induced by ischemia. However, the molecular signals leading to cyclin D/Cdk4/pRb activation following ischemic insult are presently not clear. Here, we investigate the cell division cycle 25 (Cdc25) dual-specificity phosphatases as potential upstream regulators of ischemic neuronal death and Cdk4 activation. We show that a pharmacologic inhibitor of Cdc25 family members (A, B, and C) protects mouse primary neurons from hypoxia-induced delayed death. The major contributor to the death process appears to be Cdc25A. shRNA-mediated knockdown of Cdc25A protects neurons in a delayed model of hypoxia-induced death in vitro Similar results were observed in vivo following global ischemia in the rat. In contrast, neurons singly or doubly deficient for Cdc25B/C were not significantly protective. We show that Cdc25A activity, but not level, is upregulated in vitro following hypoxia and global ischemic insult in vivo Finally, we show that shRNA targeting Cdc25A blocks Ser795 pRb phosphorylation. Overall, our results indicate a role for Cdc25A in delayed neuronal death mediated by ischemia.SIGNIFICANCE STATEMENT A major challenge in stroke is finding an effective neuroprotective strategy to treat cerebral ischemic injury. Cdc25 family member A (Cdc25A) is a phosphatase normally activated during cell division in proliferating cells. We found that Cdc25A is activated in neurons undergoing ischemic stress mediated by hypoxia in vitro and global cerebral ischemia in rats in vivo We show that pharmacologic or genetic inhibition of Cdc25A activity protects neurons from delayed death in vitro and in vivo Downregulation of Cdc25A led to reduction in retinoblastoma tumor suppressor protein (pRb) phosphorylation. An increase in pRb phosphorylation has been previously linked to ischemic neuronal death. Our results identify Cdc25A as a potential target for neuroprotectant strategy for the treatment of delayed ischemic neuronal death.


Apoptosis , Brain Ischemia/metabolism , Brain Ischemia/pathology , Neurons/metabolism , Neurons/pathology , cdc25 Phosphatases/metabolism , Animals , Cells, Cultured , Enzyme Activation , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
8.
J Biol Chem ; 290(51): 30441-52, 2015 Dec 18.
Article En | MEDLINE | ID: mdl-26538564

Emerging evidence has demonstrated a growing genetic component in Parkinson disease (PD). For instance, loss-of-function mutations in PINK1 or PARKIN can cause autosomal recessive PD. Recently, PINK1 and PARKIN have been implicated in the same signaling pathway to regulate mitochondrial clearance through recruitment of PARKIN by stabilization of PINK1 on the outer membrane of depolarized mitochondria. The precise mechanisms that govern this process remain enigmatic. In this study, we identify Bcl2-associated athanogene 2 (BAG2) as a factor that promotes mitophagy. BAG2 inhibits PINK1 degradation by blocking the ubiquitination pathway. Stabilization of PINK1 by BAG2 triggers PARKIN-mediated mitophagy and protects neurons against 1-methyl-4-phenylpyridinium-induced oxidative stress in an in vitro cell model of PD. Collectively, our findings support the notion that BAG2 is an upstream regulator of the PINK1/PARKIN signaling pathway.


Adaptor Proteins, Signal Transducing/metabolism , Mitochondria/metabolism , Molecular Chaperones/metabolism , Neurons/metabolism , Protein Kinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Survival , Mice , Mice, Mutant Strains , Mitochondria/genetics , Mitochondrial Membranes/metabolism , Mitophagy/genetics , Molecular Chaperones/genetics , Protein Kinases/genetics , Protein Stability , Protein Transport , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
Neuroreport ; 25(1): 18-22, 2014 Jan 08.
Article En | MEDLINE | ID: mdl-24296793

For many years, brain ischemia has been known to be a leading cause of adult neurological disorder. In particular, many reports have shown that hyperexcitability of neurons and inflammatory response of the glia induced by ischemic reperfusion (I/R) determine the fate of cells in the ischemic core and the penumbra region. Although there are many reports on the activation and roles of signal transducer and activator of transcription (STAT) proteins (STAT1, STAT3, and STAT5) during hyperexcitation in the neuron and inflammation occurring following I/R, the temporal and spatial activation of STAT6 protein in the ischemic cortex still remain elusive. In this study, using a transient rat middle cerebral artery occlusion model, we primarily investigated the time-course expression of the phosphorylated STAT6 (pSTAT6) in the ischemic core region following I/R, which was compared with that of pSTAT3. We found that pSTAT6 significantly decreases at 1 and 12 h following I/R, whereas pSTAT3 markedly increases at each follow-up time point. In addition, the level of pSTAT6 is reduced in the ischemic core in comparison with the penumbra region at 12 h following I/R. However, there is no significant difference in pSTAT3 expression between the ischemic core and the penumbra. Taken together, our data suggest that pSTAT6 and pSTAT3 are modulated differently following I/R during ischemic stroke.


Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Infarction, Middle Cerebral Artery/metabolism , Reperfusion , STAT6 Transcription Factor/metabolism , Animals , Cerebral Cortex/blood supply , Male , Neurons/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Time Factors
10.
Brain Res ; 1455: 114-23, 2012 May 21.
Article En | MEDLINE | ID: mdl-22483792

Excess activation of ionotropic glutamate receptors and iron is believed to contribute to free radical production and neuronal death following hypoxic ischemia. We examined the possibility that both NMDA receptor activation and iron overload determine spatial and temporal patterns of free radical production after transient middle cerebral artery occlusion (tMCAO) in male Sprague-Dawley rats. Mitochondrial free radical (MFR) levels were maximally increased in neurons in the core at 1 h and 24 h after tMCAO. Early MFR production was blocked by administration of MK-801, an NMDA receptor antagonist, but not deferoxamine, an iron chelator. Neither MK-801 nor deferoxamine attenuated late MFR production in the core. Increased MFRs were observed in penumbral neurons within 6 h and gradually increased over 24 h after tMCAO. Slowly-evolving MFRs in the core and penumbra were accompanied by iron overload. Deferoxamine blocked iron overload but reduced MFR production only in the penumbra. Combined MK-801/deferoxamine reduced late MFR production in both core and penumbra in an additive manner. Combination therapy significantly ameliorated infarction compared with monotherapy. These findings suggest that the NMDA receptor activation and iron overload mediate late MFR production and infarction after tMCAO.


Brain Damage, Chronic/metabolism , Free Radicals/metabolism , Infarction, Middle Cerebral Artery/metabolism , Iron/metabolism , Oxidative Stress/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Brain Damage, Chronic/drug therapy , Brain Damage, Chronic/physiopathology , Deferoxamine/pharmacology , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
11.
J Neurochem ; 122(5): 952-61, 2012 Sep.
Article En | MEDLINE | ID: mdl-22537108

While free radicals and inflammation constitute major routes of neuronal injury occurring in amyotrophic lateral sclerosis (ALS), neither antioxidants nor non-steroidal anti-inflammatory drugs have shown significant efficacy in human clinical trials. We examined the possibility that concurrent blockade of free radicals and prostaglandin E(2) (PGE(2))-mediated inflammation might constitute a safe and effective therapeutic approach to ALS. We have developed 2-hydroxy-5-[2-(4-trifluoromethylphenyl)-ethylaminobenzoic acid] (AAD-2004) as a derivative of aspirin. AAD-2004 completely removed free radicals at 50 nM as a potent spin-trapping molecule and inhibited microsomal PGE(2) synthase-1 (mPGES-1) activity in response to both lipopolysaccharide-treated BV2 cell with IC(50) of 230 nM and recombinant human mPGES-1 protein with IC(50) of 249 nM in vitro. In superoxide dismutase 1(G93A) transgenic mouse model of ALS, AAD-2004 blocked free radical production, PGE(2) formation, and microglial activation in the spinal cords. As a consequence, AAD-2004 reduced autophagosome formation, axonopathy, and motor neuron degeneration, improving motor function and increasing life span. In these assays, AAD-2004 was superior to riluzole or ibuprofen. Gastric bleeding was not induced by AAD-2004 even at a dose 400-fold higher than that required to obtain maximal therapeutic efficacy in superoxide dismutase 1(G93A). Targeting both mPGES-1-mediated PGE(2) and free radicals may be a promising approach to reduce neurodegeneration in ALS and possibly other neurodegenerative diseases.


Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Dinoprostone/metabolism , Free Radicals/metabolism , Sulfasalazine/therapeutic use , 8-Hydroxy-2'-Deoxyguanosine , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Analysis of Variance , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/analogs & derivatives , Aspirin/pharmacology , Aspirin/therapeutic use , Calcium-Binding Proteins/metabolism , Cerebral Cortex/pathology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/drug therapy , Free Radical Scavengers/metabolism , Free Radicals/antagonists & inhibitors , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Ibuprofen/pharmacology , Ibuprofen/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Motor Neurons/drug effects , Motor Neurons/pathology , Oxidative Stress/drug effects , Riluzole/pharmacology , Riluzole/therapeutic use , Spinal Cord/pathology , Sulfasalazine/pharmacology , Superoxide Dismutase/genetics , Tyrosine/analogs & derivatives , Tyrosine/metabolism
12.
Neurosci Lett ; 489(2): 84-8, 2011 Feb 04.
Article En | MEDLINE | ID: mdl-21134418

The purpose of the present study was to evaluate whether chronic cerebral hypoperfusion would affect cognitive status in an Alzheimer mouse model. Behavioral tests and histological evaluations were performed using female Tg2576 mice eight weeks after right common carotid artery occlusion (rCCAO), which is known to induce a type of vascular dementia without neuronal necrosis in nontransgenic mice. Positron emission tomography with (18)F-fluorodeoxyglucose (FDG-PET) was utilized to evaluate metabolic status in the rCCAO-operated brain of nontransgenic mice. Escape latency from the Morris water maze test was not significantly different between rCCAO- and sham-operated mice. However, the learning curve was impaired in rCCAO-operated transgenic mice while it was preserved in sham-operated transgenic or rCCAO-operated nontransgenic mice. Histological examination revealed no evidence of cell death in the rCCAO-operated brains, and the extent of amyloid deposition was not different in rCCAO- and sham-operated mice. The brain of rCCAO-operated mice showed metabolic deficits in the ipsilateral parietal cortex through FDG-PET. In conclusion, further cognitive decline which is more comparable to typical Alzheimer's disease was induced by chronic cerebral hypoperfusion in an Alzheimer mouse model. This aggravation might be associated with hypometabolism via chronic cerebral hypoperfusion.


Alzheimer Disease/physiopathology , Brain Ischemia/complications , Cognition Disorders/physiopathology , Alzheimer Disease/etiology , Alzheimer Disease/psychology , Animals , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Ischemia/physiopathology , Carotid Stenosis/complications , Cerebrovascular Circulation , Cognition Disorders/etiology , Cognition Disorders/psychology , Dementia, Vascular/etiology , Dementia, Vascular/physiopathology , Dementia, Vascular/psychology , Female , Glucose/metabolism , Maze Learning , Memory, Short-Term , Mice , Mice, Inbred C57BL , Mice, Transgenic , Species Specificity
13.
J Neurochem ; 114(1): 160-70, 2010 Jul.
Article En | MEDLINE | ID: mdl-20374429

Alternative splicing of tau exon 10 influences microtubule assembly and stability during development and in pathological processes of the central nervous system. However, the cellular events that underlie this pre-mRNA splicing remain to be delineated. In this study, we examined the possibility that ischemic injury, known to change the cellular distribution and expression of several RNA splicing factors, alters the splicing of tau exon 10. Transient occlusion of the middle cerebral artery reduced tau exon 10 inclusion in the ischemic cortical area within 12 h, resulting in the induction of three-repeat (3R) tau in cortical neurons. Ubiquitinated protein aggregates and reduced proteasome activity were also observed. Administration of proteasome inhibitors such as MG132, proteasome inhibitor I and lactacystin reduced tau exon 10 splicing in cortical cell cultures. Decreased levels of Tra2beta, an RNA splicing factor responsible for tau exon 10 inclusion, were detected both in cortical cell cultures exposed to MG132 and in cerebral cortex after ischemic injury. Taken together, these findings suggest that transient focal cerebral ischemia reduces tau exon 10 splicing through a mechanism involving proteasome-ubiquitin dysfunction and down-regulation of Tra2beta.


Hypoxia-Ischemia, Brain/metabolism , Ischemic Attack, Transient/metabolism , Proteasome Endopeptidase Complex/physiology , tau Proteins/metabolism , Alternative Splicing , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Exons , Male , Neurons/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Proteasome Inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Serine-Arginine Splicing Factors , Ubiquitination , tau Proteins/genetics
14.
J Cereb Blood Flow Metab ; 27(6): 1142-51, 2007 Jun.
Article En | MEDLINE | ID: mdl-17106444

Excitotoxicity and oxidative stress mediate neuronal death after hypoxic-ischemic brain injury. We examined the possibility that targeting both N-methyl-D-aspartate (NMDA) receptor-mediated excitotoxicity and oxidative stress would result in enhanced neuroprotection against hypoxic-ischemia. 2-Hydroxy-5-(2,3,5,6-tetrafluoro-4-trifluoromethyl-benzylamino)-benzoic acid (Neu2000) was derived from aspirin and sulfasalazine to prevent both NMDA neurotoxicity and oxidative stress. In cortical cell cultures, Neu2000 was shown to be an uncompetitive NMDA receptor antagonist and completely blocked free radical toxicity at doses as low as 0.3 micromol/L. Neu2000 showed marked neuroprotection in a masked fashion using histology and behavioral testing in two rodent models of focal cerebral ischemia without causing neurotoxic side effects. Neu2000 protected against the effects of middle cerebral artery occlusion, even when delivered 8 h after reperfusion. Single bolus administration of the drug prevented gray and white matter degeneration and spared neurologic function for over 28 days after MACO. Neu2000 may be a novel therapy for combating both NMDA receptor-mediated excitotoxicity and oxidative stress, the two major routes of neuronal death in ischemia, offering profound neuroprotection and an extended therapeutic window.


Antioxidants/pharmacology , Benzoates/pharmacology , Brain Ischemia/prevention & control , N-Methylaspartate/antagonists & inhibitors , Animals , Aspirin/chemistry , Benzoates/therapeutic use , Brain Ischemia/drug therapy , Cells, Cultured , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/pharmacology , Fluorobenzenes , Infarction, Middle Cerebral Artery , Mice , Oxidative Stress/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Salicylates , Sulfasalazine/chemistry , meta-Aminobenzoates
...