Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Exp Cell Res ; 315(16): 2856-68, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19559021

ABSTRACT

Collapsin response mediator proteins (CRMPs) are key modulators of cytoskeletons during neurite outgrowth in response to chemorepulsive guidance molecules. However, their roles in adult injured neurons are not well understood. We previously demonstrated that CRMP3 underwent calcium-dependent N-terminal protein cleavage during excitotoxicity-induced neurite retraction and neuronal death. Here, we report findings that the full-length CRMP3 inhibits tubulin polymerization and neurite outgrowth in cultured mature cerebellar granule neurons, while the N-terminal truncated CRMP3 underwent nuclear translocation and caused a significant nuclear condensation. The N-terminal truncated CRMP3 underwent nuclear translocation through nuclear pores. Nuclear protein pull-down assay and mass spectrometry analysis showed that the N-terminal truncated CRMP3 was associated with nuclear vimentin. In fact, nuclear-localized CRMP3 co-localized with vimentin during glutamate-induced excitotoxicity. However, the association between the truncated CRMP3 and vimentin was not critical for nuclear condensation and neurite outgrowth since over-expression of truncated CRMP3 in vimentin null neurons did not alleviate nuclear condensation and neurite outgrowth inhibition. Together, these studies showed CRMP3's role in attenuating neurite outgrowth possibility through inhibiting microtubule polymerization, and also revealed its novel association with vimentin during nuclear condensation prior to neuronal death.


Subject(s)
Calpain/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Protein Isoforms/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Humans , Mice , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Protein Isoforms/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vimentin/metabolism
2.
Eur J Neurosci ; 26(4): 801-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17672855

ABSTRACT

Collapsin response mediator proteins (CRMPs) are important brain-specific proteins with distinct functions in modulating growth cone collapse and axonal guidance during brain development. Our previous studies have shown that calpain cleaves CRMP3 in the adult mouse brain during cerebral ischemia [S.T. Hou et al. (2006) J. Neurosci., 26, 2241-2249]. Here, the expression of all CRMP family members (1-5) was examined in mouse brains that were subjected to middle cerebral artery occlusion. Among the five CRMPs, the expressions of CRMP1, CRMP3 and CRMP5 were the most abundant in the cerebral cortex and all CRMPs were targeted for cleavage by ischemia-activated calpain. Sub-cellular fractionation analysis showed that cleavage of CRMPs by calpain occurred not only in the cytoplasm but also in the synaptosomes isolated from ischemic brains. Moreover, synaptosomal CRMPs appeared to be at least one-fold more sensitive to cleavage compared with those isolated from the cytosolic fraction in an in-vitro experiment, suggesting that synaptosomal CRMPs are critical targets during cerebral ischemia-induced neuronal injury. Finally, the expression of all CRMPs was colocalized with TUNEL-positive neurons in the ischemic mouse brain, which further supports the notion that CRMPs may play an important role in neuronal death following cerebral ischemia. Collectively, these studies demonstrated that CRMPs are targets of calpains during cerebral ischemia and they also highlighted an important potential role that CRMPs may play in modulating ischemic neuronal death.


Subject(s)
Amidohydrolases/metabolism , Brain Ischemia/metabolism , Calpain/metabolism , Nerve Tissue Proteins/metabolism , Animals , Blotting, Western , Cell Death/physiology , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiology , Cytoplasmic Granules/physiology , Data Interpretation, Statistical , Hydrolases , Immunohistochemistry , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins , Neurons/physiology , Subcellular Fractions/metabolism , Synaptosomes/metabolism
3.
Neurosci Lett ; 419(1): 23-7, 2007 May 23.
Article in English | MEDLINE | ID: mdl-17418946

ABSTRACT

Selective gene expression targeting neurons is a challenge, which, if successfully overcome, carries an enormous potential for clinical applications in therapeutics against neurodegenerative diseases. We have reported previously the construction of a series of adenoviral vectors capable of selectively expressing a reporter gene luciferase in cultured neurons [D. Huang, A. Desbois, S.T. Hou, A novel adenoviral vector which mediates hypoxia-inducible gene expression selectively in neurons, Gene Ther. 12 (2005) 1369-1376]. A combination of neuron restrictive silencer elements (NRSEs), hypoxia responsive elements (HREs) and CMV minimal promoter (CMVmp) was packaged into replication defective adenovirus to target gene expression selectively in neurons in a hypoxia-regulated manner. In the present study, we injected the adenoviral vectors into the neonatal mouse brain followed by treatment with hypoxia. The expression of the reporter luciferase gene was examined by luciferase assay and fluorescent immunostaining. Neurons and glial cells were identified by staining with antibodies against NeuN and GFAP, respectively. Remarkably, in response to hypoxia, Ad/5HRE-3NRSE showed strong hypoxia-inducible gene expression of the reporter luciferase selectively in neurons but not in glial cells. In contrast, brains infected with the control vector Ad/5HRE showed no selectivity in luciferase expression (in both neurons and glial cells) under the hypoxic condition. Taken together, these studies demonstrated that this vector (Ad/5HRE-3NRSE) can mediate gene expression selectively in neurons both in vitro and in vivo, supporting the suggestion that further refinement of this vector may lead to the development of a useful tool to investigate mechanisms of neuronal damage following cerebral ischemia and a possible effective gene therapy vector to stroke.


Subject(s)
Brain/metabolism , Cytomegalovirus/physiology , Gene Expression/physiology , Genetic Vectors/administration & dosage , Hypoxia/pathology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Line, Transformed , Gene Transfer Techniques , Genetic Vectors/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Hypoxia/physiopathology , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Phosphopyruvate Hydratase/metabolism
4.
Mol Cell Biol ; 27(5): 1696-705, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17178835

ABSTRACT

The nuclear transcription factor E2F1 plays an important role in modulating neuronal death in response to excitotoxicity and cerebral ischemia. Here, by comparing gene expression in brain cortices from E2F1(+/+) and E2F1(-/-) mice using a custom high-density DNA microarray, we identified a group of putative E2F1 target genes that might be responsible for ischemia-induced E2F1-dependent neuronal death. Neuropilin 1 (NRP-1), a receptor for semaphorin 3A-mediated axon growth cone collapse and retraction, was confirmed to be a direct target of E2F1 based on (i) the fact that the NRP-1 promoter sequence contains an E2F1 binding site, (ii) reactivation of NRP-1 expression in E2F1(-/-) neurons when the E2F1 gene was replaced, (iii) activation of the NRP-1 promoter by E2F1 in a luciferase reporter assay, (iv) electrophoretic mobility gel shift analysis confirmation of the presence of an E2F binding sequence in the NRP-1 promoter, and (v) the fact that a chromatin immunoprecipitation assay showed that E2F1 binds directly to the endogenous NRP-1 promoter. Interestingly, the temporal induction in cerebral ischemia-induced E2F1 binding to the NRP-1 promoter correlated with the temporal-induction profile of NRP-1 mRNA, confirming that E2F1 positively regulates NRP-1 during cerebral ischemia. Functional analysis also showed that NRP-1 receptor expression was extremely low in E2F1(-/-) neurons, which led to the diminished response to semaphorin 3A-induced axonal shortening and neuronal death. An NRP-1 selective peptide inhibitor provided neuroprotection against oxygen-glucose deprivation. Taken together, these findings support a model in which E2F1 targets NRP-1 to modulate axonal damage and neuronal death in response to cerebral ischemia.


Subject(s)
Brain Death/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , E2F1 Transcription Factor/genetics , Neurons/metabolism , Neuropilin-1/metabolism , Adenoviridae/genetics , Animals , Brain Ischemia/etiology , Cells, Cultured , Cerebellum/cytology , Chromatin Immunoprecipitation , E2F1 Transcription Factor/metabolism , Electrophoretic Mobility Shift Assay , Genes, Reporter , Luciferases/analysis , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Biological , Neuroglia/cytology , Neurons/pathology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
5.
J Neurosci ; 26(8): 2241-9, 2006 Feb 22.
Article in English | MEDLINE | ID: mdl-16495451

ABSTRACT

Collapsin response mediator proteins (CRMPs) mediate growth cone collapse during development, but their roles in adult brains are not clear. Here we report the findings that the full-length CRMP-3 (p63) is a direct target of calpain that cleaves CRMP-3 at the N terminus (+76 amino acid). Interestingly, activated calpain in response to excitotoxicity in vitro and cerebral ischemia in vivo also cleaved CRMP-3, and the cleavage product of CRMP-3 (p54) underwent nuclear translocation during neuronal death. The expression of p54 was colocalized with the terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling-positive nuclei in glutamate-treated cerebellar granule neurons (CGNs) and in ischemic neurons located in the infarct core after focal cerebral ischemia, suggesting that p54 might be involved in neuronal death. Overexpression studies showed that p54, but not p63, caused death of human embryonic kidney cells and CGNs, whereas knock-down CRMP-3 expression by selective small interfering RNA protected neurons against glutamate toxicity. Collectively, these results reveal a novel role of CRMP-3 in that calpain cleavage of CRMP-3 and the subsequent nuclear translocation of the truncated CRMP-3 evokes neuronal death in response to excitotoxicity and cerebral ischemia. Our findings also establish a novel route of how calpain signals neuron death.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Calpain/metabolism , Glutamic Acid/toxicity , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Apoptosis/drug effects , Binding Sites , Brain/drug effects , Cells, Cultured , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/pathology , Protein Binding
6.
Brain Res Mol Brain Res ; 136(1-2): 1-11, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15893581

ABSTRACT

We isolated a fragment of the fukutin gene promoter from differentiated human NT2 cells using chromatin immunoprecipitation technique with an anti-CREB antibody. This fragment contained a CRE-like sequence and here we describe its functional validation. The results showed that the element was functional in vitro and in vivo and that CREB in neurons was involved in the transcriptional regulation of the fukutin gene. Moreover, its expression in neurons was regulated by cAMP and calcium ions, known triggers of CREB phosphorylation. To our knowledge, this is the first report on the regulation of fukutin gene by transcription factor CREB in response to the signals generated by synaptic activity. The true biological function of fukutin, the gene responsible for Fukuyama-type congenital muscular dystrophy and mental retardation, is at present not known. However, it has been suggested that it might possess glycosyltransferase activity and its intracellular localization within the Golgi structures is consistent with this function. As such, fukutin might play a significant role in post-translational modification of synaptic proteins in neuronal cells.


Subject(s)
Cyclic AMP Response Element-Binding Protein/isolation & purification , Promoter Regions, Genetic/physiology , Proteins/genetics , Autoantigens/metabolism , Blotting, Northern/methods , Blotting, Western/methods , Cell Line, Tumor , Chromatin Immunoprecipitation/methods , Cloning, Molecular , Colforsin/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Electrophoretic Mobility Shift Assay/methods , Fluorescent Antibody Technique/methods , Gene Expression Regulation/drug effects , Genes, Reporter/physiology , Golgi Matrix Proteins , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins , Potassium Chloride/pharmacology , Protein Binding , Proteins/metabolism , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Subcellular Fractions/metabolism , Teratocarcinoma , Transcriptional Activation/physiology , Transfection/methods
7.
Neurosci Lett ; 377(2): 97-100, 2005 Mar 29.
Article in English | MEDLINE | ID: mdl-15740844

ABSTRACT

The present study investigated the role of pyridoxal phosphate-6-azophenyl-2',4'-disulfonic acid tetrasodium salt (PPADS), a P2 receptor antagonist, in protecting mouse cerebellar granule neurons (CGNs) against glutamate/NMDA-induced neuronal death. Neurotoxicity caused by 50 microM glutamate or 200 microM NMDA was significantly reduced in CGNs treated with PPADS. Such neuroprotection was in a time- and dose-dependent manner. The possibility that PPADS may block glutamate/NMDA-mediated intracellular Ca2+ influx to CGNs was investigated using temperature-controlled platereader measurements of fluorescence intensity of CGNs loaded with Ca2+-sensitive fluorescent dye Fluo-4AM. Interestingly, the rapid increase of calcium influx following glutamate/NMDA treatment was not significantly affected by prior treatment with PPADS. In contrast, MK801, a specific NMDA receptor antagonist, completely blocked intracellular Ca2+ influx. Taken together, these data suggest that inhibition of the P2 receptor may directly modulate NMDA receptor-mediated neurotoxicity through a Ca2+-independent mechanism.


Subject(s)
Glutamic Acid/toxicity , N-Methylaspartate/toxicity , Neuroprotective Agents/pharmacology , Purinergic P2 Receptor Antagonists , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Receptors, Purinergic P2/physiology
8.
J Mol Neurosci ; 24(2): 323-8, 2004.
Article in English | MEDLINE | ID: mdl-15456946

ABSTRACT

Expression of therapeutic gene products in differentiated human NT2 neurons (NT2/Ns) is being explored for ex vivo gene therapy of human neurological diseases. In this study we determined the efficiency of adenovirus (Ad)-mediated gene delivery into NT2/Ns and characterized the expression of several key receptors known to be required for efficient Ad-mediated gene delivery. Undifferentiated NT2 cells and NT2/Ns were infected by Ad expressing green fluorescent protein at an efficiency of 33% and 17%, respectively percentages much lower than the 92% infectivity obtained from a human non-neuronal cell line A549 cells. This relatively low infectivity of NT2/Ns might be caused by the extremely low expression of integrin subunit beta3 and the reduced expression of beta5 during differentiation. The expression of coxsackie-Ad receptor (CAR) was relatively high and remained constant during differentiation. Blocking CAR receptor using an antibody specific against CAR reduced Ad infectivity in a dose-dependent manner. These observations suggest that modulating the expression of integrin subunits beta3/5 or the functional heterodimer alphavbeta3/5 in human NT2/Ns may enhance adenoviral infectivity of NT2/Ns.


Subject(s)
Gene Transfer Techniques , Integrin beta Chains/metabolism , Integrin beta3/metabolism , Neurons/metabolism , Receptors, Virus/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Cell Differentiation , Cell Line , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Humans , Integrin beta Chains/genetics , Integrin beta3/genetics , Neurons/cytology , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Virus/genetics
9.
Neuroreport ; 15(14): 2241-4, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15371742

ABSTRACT

The role of B group vitamins in preventing neuronal death against excitotoxicity was investigated. Neuronal death of cultured mouse cerebellar granule neurons (CGNs) caused by glutamate (50 microM) or NMDA (200 microM) was delayed in CGNs that had been treated with riboflavin (B2), folic acid (B9) or cynocobalamin (B12) for 18 h. Such neuroprotection by B2, B9 and B12 was in a dose- and time-dependent manner. In contrast, application of thiamin (B1), nicotinamide (B3), d-pantothenic acid (B5), pyridoxine (B6) or carnitine (BT) did not ameliorate glutamate or NMDA-mediated excitotoxicity to CGCs. These results are the first indication that certain B group vitamins are not only required for the normal brain function, but can also play a protective role against excitotoxicity to the brain.


Subject(s)
Cerebellum/drug effects , Glutamic Acid/toxicity , N-Methylaspartate/toxicity , Neuroprotective Agents/pharmacology , Vitamin B Complex/pharmacology , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Cerebellum/pathology , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/pathology , Riboflavin/pharmacology , Vitamin B 12/pharmacology
10.
J Biol Chem ; 278(29): 26401-9, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12738784

ABSTRACT

We have identified a functional cAMP-response element (CRE) in the human brain-derived neurotrophic factor (BDNF) gene promoter III and established that it participated in the modulation of BDNF expression in NT2/N neurons via downstream signaling from the D1 class of dopamine (DA) receptors. The up-regulation of BDNF expression, in turn, produced neuroprotective signals through receptor tyrosine kinase B (TrkB) and promoted cell survival under the conditions of oxygen and glucose deprivation. To our knowledge this is the first evidence showing the presence of a functional CRE in the human BDNF gene and the role of DA signaling in establishing transcriptional competence of CRE in post-mitotic NT2/N neurons. This ability of DA to regulate the expression of the BDNF survival factor has a profound significance for the nigrostriatal pathway, because it indicates the existence of a feedback loop between the neutrophin, which promotes both the maturation and survival of dopaminergic neurons, and the neurotransmitter, which the mature neurons ultimately produce and release.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Dopamine/metabolism , Promoter Regions, Genetic , Animals , Base Sequence , Bucladesine/pharmacology , Cell Differentiation , Cell Line , Cell Survival , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA/genetics , DNA/metabolism , Humans , Molecular Sequence Data , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Receptor, trkB/metabolism , Receptors, Dopamine D1/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Signal Transduction , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...