Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 291(53): 27007-27022, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27864367

ABSTRACT

Phaseic acid (PA) is a phytohormone regulating important physiological functions in higher plants. Here, we show the presence of naturally occurring (-)-PA in mouse and rat brains. (-)-PA is exclusively present in the choroid plexus and the cerebral vascular endothelial cells. Purified (-)-PA has no toxicity and protects cultured cortical neurons against glutamate toxicity through reversible inhibition of glutamate receptors. Focal occlusion of the middle cerebral artery elicited a significant induction in (-)-PA expression in the cerebrospinal fluid but not in the peripheral blood. Importantly, (-)-PA induction only occurred in the penumbra area, indicting a protective role of PA in the brain. Indeed, elevating the (-)-PA level in the brain reduced ischemic brain injury, whereas reducing the (-)-PA level using a monoclonal antibody against (-)-PA increased ischemic injury. Collectively, these studies showed for the first time that (-)-PA is an endogenous neuroprotective molecule capable of reversibly inhibiting glutamate receptors during ischemic brain injury.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Glutamic Acid/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Receptors, Glutamate/chemistry , Sesquiterpenes/therapeutic use , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/etiology , Brain Ischemia/metabolism , Cells, Cultured , Infarction, Middle Cerebral Artery/etiology , Infarction, Middle Cerebral Artery/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism
2.
Sci Rep ; 5: 7890, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25601765

ABSTRACT

Semaphorin 3A (Sema3A) increased significantly in mouse brain following cerebral ischemia. However, the role of Sema3A in stroke brain remains unknown. Our aim was to determine wether Sema3A functions as a vascular permeability factor and contributes to ischemic brain damage. Recombinant Sema3A injected intradermally to mouse skin, or stereotactically into the cerebral cortex, caused dose- and time-dependent increases in vascular permeability, with a degree comparable to that caused by injection of a known vascular permeability factor vascular endothelial growth factor receptors (VEGF). Application of Sema3A to cultured endothelial cells caused disorganization of F-actin stress fibre bundles and increased endothelial monolayer permeability, confirming Sema3A as a permeability factor. Sema3A-mediated F-actin changes in endothelial cells were through binding to the neuropilin2/VEGFR1 receptor complex, which in turn directly activates Mical2, a F-actin modulator. Down-regulation of Mical2, using specific siRNA, alleviated Sema3A-induced F-actin disorganization, cellular morphology changes and endothelial permeability. Importantly, ablation of Sema3A expression, cerebrovascular permeability and brain damage were significantly reduced in response to transient middle cerebral artery occlusion (tMCAO) and in a mouse model of cerebral ischemia/haemorrhagic transformation. Together, these studies demonstrated that Sema3A is a key mediator of cerebrovascular permeability and contributes to brain damage caused by cerebral ischemia.


Subject(s)
Brain Injuries/genetics , Capillary Permeability/genetics , Semaphorin-3A/genetics , Stroke/genetics , Actins/genetics , Actins/metabolism , Animals , Apoptosis/genetics , Brain Injuries/physiopathology , Brain Ischemia/genetics , Brain Ischemia/physiopathology , Capillary Permeability/physiology , Cerebral Cortex/injuries , Cerebral Cortex/pathology , Cytoskeletal Proteins/biosynthesis , Disease Models, Animal , Humans , Mice , Microfilament Proteins , Neuropilin-2/metabolism , Semaphorin-3A/administration & dosage , Stroke/physiopathology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
3.
PLoS One ; 8(5): e64894, 2013.
Article in English | MEDLINE | ID: mdl-23741413

ABSTRACT

Excessive activation of the N-methyl-D-aspartic acid (NMDA) type glutamate receptors (NMDARs) causes excitotoxicity, a process important in stroke-induced neuronal death. Drugs that inhibit NMDA receptor-mediated [Ca(2+)]i influx are potential leads for development to treat excitotoxicity-induced brain damage. Our previous studies showed that 2-(2-benzofu-ranyl)-2-imidazoline (2-BFI), an immidazoline receptor ligand, dose-dependently protects rodent brains from cerebral ischemia injury. However, the molecular mechanisms remain unclear. In this study, we found that 2-BFI transiently and reversibly inhibits NMDA, but not AMPA currents, in a dose-dependent manner in cultured rat cortical neurons. The mechanism of 2-BFI inhibition of NMDAR is through a noncompetitive fashion with a faster on (Kon = 2.19±0.33×10(-9) M(-1) sec(-1)) and off rate (Koff = 0.67±0.02 sec(-1)) than those of memantine, a gold standard for therapeutic inhibition NMDAR-induced excitotoxicity. 2-BFI also transiently and reversibly blocked NMDA receptor-mediated calcium entry to cultured neurons and provided long-term neuroprotection against NMDA toxicity in vitro. Collectively, these studies demonstrated a potential mechanism of 2-BFI-mediated neuroprotection and indicated that 2-BFI is an excellent candidate for repositioning as a drug for stroke treatment.


Subject(s)
Benzofurans/pharmacology , Imidazoles/pharmacology , Membrane Potentials/drug effects , N-Methylaspartate/pharmacology , Neuroprotective Agents/pharmacology , Animals , Benzofurans/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Imidazoles/metabolism , Kinetics , Male , N-Methylaspartate/metabolism , N-Methylaspartate/toxicity , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/metabolism , Patch-Clamp Techniques , Protein Binding , Rats , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism
4.
Sci Rep ; 3: 1350, 2013.
Article in English | MEDLINE | ID: mdl-23443259

ABSTRACT

CRMP proteins play critical regulatory roles during semaphorin-mediated neurite outgrowth, neuronal differentiation and death. Albeit having a high degree of structure and sequence resemblance to that of liver dihydropyrimidinase, purified rodent brain CRMPs do not hydrolyze dihydropyrimidinase substrates. Here we found that mouse CRMP3 has robust histone H4 deacetylase activity. During excitotoxicity-induced mouse neuronal death, calpain-cleaved, N-terminally truncated CRMP3 undergoes nuclear translocation to cause nuclear condensation through deacetylation of histone H4. CRMP3-mediated deacetylation of H4 leads to de-repression of the E2F1 gene transcription and E2F1-dependent neuronal death. These studies revealed a novel mechanism of CRMP3 in neuronal death. Together with previous well established bodies of literature that inhibition of histone deacetylase activity provides neuroprotection, we envisage that inhibition of CRMP3 may represent a novel therapeutic approach towards excitotoxicity-induced neuronal death.


Subject(s)
Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Histones/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Acetylation , Animals , Cell Death/drug effects , Cell Death/genetics , Cell Line , E2F1 Transcription Factor/genetics , Female , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glutamic Acid/pharmacology , Histone Deacetylases/metabolism , Humans , Mice , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/ultrastructure , Protein Binding , Protein Transport/drug effects , Tubulin
5.
J Neurochem ; 122(4): 764-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22681613

ABSTRACT

Microglia are the 'immune cells' of the brain and their activation plays a vital role in the pathogenesis of many neurodegenerative diseases. Activated microglia produce high levels of pro-inflammatory factors, such as TNFα, causing neurotoxicity. Here we show that vimentin played a key role in controlling microglia activation and neurotoxicity during cerebral ischemia. Deletion of vimentin expression significantly impaired microglia activation in response to LPS in vitro and transient focal cerebral ischemia in vivo. Reintroduction of the functional vimentin gene back into vimentin knockout microglia restored their response to LPS. More importantly, impairment of microglia activation significantly protected brain from cerebral ischemia-induced neurotoxicity. Collectively, we demonstrate a previously unknown function of vimentin in controlling microglia activation.


Subject(s)
Brain Ischemia/pathology , Macrophage Activation/physiology , Microglia/physiology , Vimentin/physiology , Animals , Blotting, Western , Brain/pathology , Cell Separation , Fluorescent Antibody Technique, Indirect , Image Processing, Computer-Assisted , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/pathology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microscopy, Confocal , Nervous System Diseases/genetics , Nervous System Diseases/physiopathology , Plasmids/drug effects , Plasmids/genetics , Reperfusion Injury/pathology , Tetracycline/pharmacology , Vimentin/genetics
6.
Mol Biol Cell ; 23(4): 553-66, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22190742

ABSTRACT

Dystonin/Bpag1 is a cytoskeletal linker protein whose loss of function in dystonia musculorum (dt) mice results in hereditary sensory neuropathy. Although loss of expression of neuronal dystonin isoforms (dystonin-a1/dystonin-a2) is sufficient to cause dt pathogenesis, the diverging function of each isoform and what pathological mechanisms are activated upon their loss remains unclear. Here we show that dt(27) mice manifest ultrastructural defects at the endoplasmic reticulum (ER) in sensory neurons corresponding to in vivo induction of ER stress proteins. ER stress subsequently leads to sensory neurodegeneration through induction of a proapoptotic caspase cascade. dt sensory neurons display neurodegenerative pathologies, including Ca(2+) dyshomeostasis, unfolded protein response (UPR) induction, caspase activation, and apoptosis. Isoform-specific loss-of-function analysis attributes these neurodegenerative pathologies to specific loss of dystonin-a2. Inhibition of either UPR or caspase signaling promotes the viability of cells deficient in dystonin. This study provides insight into the mechanism of dt neuropathology and proposes a role for dystonin-a2 as a mediator of normal ER structure and function.


Subject(s)
Carrier Proteins/physiology , Cytoskeletal Proteins/physiology , Endoplasmic Reticulum/physiology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Animals , Apoptosis , Calcium/metabolism , Carrier Proteins/genetics , Caspases/biosynthesis , Cytoskeletal Proteins/genetics , Dystonia Musculorum Deformans/genetics , Dystonia Musculorum Deformans/metabolism , Dystonia Musculorum Deformans/pathology , Dystonin , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Stress , Enzyme Activation , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Neurons/pathology , Protein Isoforms/genetics , Protein Isoforms/physiology , Unfolded Protein Response
7.
Biochem Biophys Res Commun ; 401(3): 435-9, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20869945

ABSTRACT

Brain microglia are resident macrophage-like cells representing the first and main form of active immune response during brain injury. Microglia-mediated inflammatory events in the brain are known to be associated with chronic degenerative diseases such as Multiple Sclerosis, Parkinson's, or Alzheimer's disease. Therefore, identification of mechanisms activating microglia is not only important in the understanding of microglia-mediated brain pathologies, but may also lead to the development of new anti-inflammatory drugs for the treatment of chronic neurodegenerative diseases. Recently, abscisic acid (ABA), a phytohormone regulating important physiological functions in higher plants, has been proposed to activate murine microglial cell line N9 through increased intracellular calcium. In the present study, we determined the response to ABA and its analogues from murine primary microglia and immortalized murine microglial cell line BV-2 and N9 cells. A Fura-2-acetoxymethyl ester (Fura-2AM)-based ratiometric calcium imaging and measurement technique was used to determine the intracellular calcium changes in these cells when treated with (-)-ABA, (+)-ABA, (-)-trans-ABA and (+)-trans-ABA. Both primary microglia and microglial cell lines (BV-2 and N9 cells) showed significant increase in intracellular calcium ([Ca(2+)]i) in response to treatment with ATP and ionomycine. However, ABAs failed to evoke dose- and time-dependent [Ca(2+)]i changes in mouse primary microglia, BV-2 and N9 cells. Together, these surprising findings demonstrate that, contrary to that reported in N9 cells [3], ABAs do not evoke intracellular calcium changes in primary microglia and microglial cell lines. The broad conclusion that ABA evokes [Ca(2+)]i in microglia requires more evidence and further careful examination.


Subject(s)
Abscisic Acid/analogs & derivatives , Abscisic Acid/pharmacology , Calcium/metabolism , Microglia/drug effects , Animals , Cell Line, Transformed , Mice , Microglia/metabolism
8.
J Biol Chem ; 285(13): 9908-9918, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20133938

ABSTRACT

Neuropilins (NRPs) are receptors for the major chemorepulsive axonal guidance cue semaphorins (Sema). The interaction of Sema3A/NRP1 during development leads to the collapse of growth cones. Here we show that Sema3A also induces death of cultured cortical neurons through NRP1. A specific NRP1 inhibitory peptide ameliorated Sema3A-evoked cortical axonal retraction and neuronal death. Moreover, Sema3A was also involved in cerebral ischemia-induced neuronal death. Expression levels of Sema3A and NRP1, but not NRP2, were significantly increased early during brain reperfusion following transient focal cerebral ischemia. NRP1 inhibitory peptide delivered to the ischemic brain was potently neuroprotective and prevented the loss of motor functions in mice. The integrity of the injected NRP1 inhibitory peptide into the brain remained unchanged, and the intact peptide permeated the ischemic hemisphere of the brain as determined using MALDI-MS-based imaging. Mechanistically, NRP1-mediated axonal collapse and neuronal death is through direct and selective interaction with the cytoplasmic tyrosine kinase Fer. Fer RNA interference effectively attenuated Sema3A-induced neurite retraction and neuronal death in cortical neurons. More importantly, down-regulation of Fer expression using Fer-specific RNA interference attenuated cerebral ischemia-induced brain damage. Together, these studies revealed a previously unknown function of NRP1 in signaling Sema3A-evoked neuronal death through Fer in cortical neurons.


Subject(s)
Neuropilin-1/metabolism , Protein-Tyrosine Kinases/metabolism , Semaphorin-3A/chemistry , Animals , Brain/metabolism , Brain Ischemia/pathology , Cell Death , Gene Expression Regulation, Enzymologic , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neuropilin-1/chemistry , Peptides/chemistry , Protein Binding , RNA Interference , Signal Transduction
9.
Eur J Pharmacol ; 629(1-3): 12-9, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-19958763

ABSTRACT

Intracellular calcium ([Ca(2+)]i) influx through N-methyl-d-aspartic acid (NMDA) receptors in cortical neurons is central to NMDA receptor-mediated excitotoxicity. Drugs that uncompetitively modulate NMDA receptor-mediated [Ca(2+)]i influx are potential leads for development to treat NMDA receptor-mediated neuronal damage since these drugs spare NMDA receptor normal functions. Ligands to alpha(2)-adrenoceptors and imidazoline I(2) receptors confer neuroprotection possibility through modulating NMDA receptor-mediated [Ca(2+)]i influx. Here, we investigated the characteristics of several ligands to alpha(2)-adrenoceptors and imidazoline I(2) receptor, in inhibiting NMDA receptor-mediated [Ca(2+)]i influx in cultured cortical neurons using a ratiometric calcium imaging technique. In contrast to MK801, which non-reversibly blocks NMDA receptor-mediated [Ca(2+)]i influx, imidazoline I(2) receptor antagonists, Idazoxan, and 2-(2-benzofuranyl)-2-imidazoline (2-BFI)-mediated inhibition of [Ca(2+)]i influx can be rapidly reversed when removed, in a manner similar to that of memantine, an uncompetitive antagonist to NMDA receptors. Interestingly, ligands to alpha(2)-adrenoceptors, including agmatine sulfate and yohimbine, and a ligand to the nicotinic receptor, levamisol, neither inhibited NMDA receptor-mediated [Ca(2+)]i influx, nor provided neuroprotection against glutamate toxicity, suggesting selective inhibition of NMDA receptor activities. The inhibition of NMDA receptor by Idazoxan and 2-BFI also led to the suppression of NMDA receptor-mediated calpain activity as a result of blocking NMDA receptor activity, rather than through direct inhibition of calpain activity. Collectively, these studies demonstrated that imidazoline I(2) receptor antagonists transiently and reversibly block NMDA receptor-mediated [Ca(2+)]i influx. These compounds are leads for further development as uncompetitive antagonists to NMDA receptor-mediated excitotoxicity.


Subject(s)
Calcium/metabolism , Imidazoline Receptors/antagonists & inhibitors , Intracellular Space/drug effects , Intracellular Space/metabolism , Neuroprotective Agents/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Benzofurans/pharmacology , Cerebral Cortex/cytology , Glutamic Acid/toxicity , Idazoxan/pharmacology , Imidazoles/pharmacology , Mice , Molecular Imaging , Neurons/drug effects , Neurons/metabolism , Neurotoxins/toxicity
10.
Neurosci Res ; 66(4): 396-401, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20036291

ABSTRACT

Neuropilin 2 (NRP2) is a type I transmembrane protein that binds to distinct members of the class III secreted Semaphorin subfamily. NRP2 plays important roles in repulsive axon guidance, angiogenesis and vasculogenesis through partnering with co-receptors such as vascular endothelial growth factor receptors (VEGFRs) during development. Emerging evidence also suggests that NRP2 contributes to injury response and environment changes in adult brains. In this study, we examined the contribution of NRP2 gene to cerebral ischemia-induced brain injury using NRP2 deficient mouse. To our surprise, the lack of NRP2 expression does not affect the outcome of brain injury induced by transient occlusion of the middle cerebral artery (MCAO) in mouse. The cerebral vasculature in terms of the middle cerebral artery anatomy and microvessel density in the cerebral cortex of NRP2 deficient homozygous (NRP2(-/-)) mice are normal and almost identical to those of the heterozygous (NRP2(+/-)) and wild type (NRP2(+/+)) littermates. MCAO (1h) and 24h reperfusion caused a brain infarction of 23% (compared to the contralateral side) in NRP2(-/-) mice, which is not different from those in NRP2(+/- and +/+) mice at 22 and 21%, respectively (n=19, p>0.05). Correspondingly, NRP2(-/-) mouse also showed a similar level of deterioration of neurological functions after stroke compared with their NRP2(+/- and +/+) littermates. Oxygen-glucose-deprivation (OGD) caused a significant neuronal death in NRP2(-/-) cortical neurons, at the level similar to that in NRP(+/+) cortical neurons (72% death in NRP(-/-) neurons vs. 75% death in NRP2(+/+) neurons; n=4; p>0.05). Together, these loss-of-function studies demonstrated that despite of its critical role in neuronal guidance and vascular formation during development, NRP2 expression dose not affect adult brain response to cerebral ischemia.


Subject(s)
Cerebral Cortex/pathology , Glucose/deficiency , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/pathology , Neurons/pathology , Neuropilin-2/genetics , Oxygen/physiology , Animals , Brain Infarction/etiology , Brain Infarction/metabolism , Brain Infarction/pathology , Cell Survival , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/metabolism , Ischemic Attack, Transient/etiology , Ischemic Attack, Transient/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Neuropilin-2/biosynthesis
11.
J Neurochem ; 111(3): 870-81, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19735446

ABSTRACT

Intracellular calcium influx through NMDA receptors triggers a cascade of deleterious signaling events which lead to neuronal death in neurological conditions such as stroke. However, it is not clear as to the molecular mechanism underlying early damage response from axons and dendrites which are important in maintaining a network essential for the survival of neurons. Here, we examined changes of axons treated with glutamate and showed the appearance of betaIII-tubulin positive varicosities on axons before the appearance of neuronal death. Dizocilpine blocked the occurrence of varicosities on axons suggesting that these microstructures were mediated by NMDA receptor activities. Despite early increased expression of pCaMKII and pMAPK after just 10 min of glutamate treatment, only inhibitors to Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and calpain prevented the occurrence of axonal varicosities. In contrast, inhibitors to Rho kinase, mitogen-activated protein kinase and phosphoinositide 3-kinase were not effective, nor were they able to rescue neurons from death, suggesting CaMKII and calpain are important in axon survival. Activated CaMKII directly phosphorylates collapsin response mediator protein (CRMP) 2 which is independent of calpain-mediated cleavage of CRMP2. Over-expression of CRMP2, but not the phosphorylation-resistant mutant CRMP2-T555A, increased axonal resistance to glutamate toxicity with reduced numbers of varicosities. The levels of both pCRMP2 and pCaMKII were also increased robustly within early time points in ischemic brains and which correlated with the appearance of axonal varicosities in the ischemic neurons. Collectively, these studies demonstrated an important role for CaMKII in modulating the integrity of axons through CRMP2 during excitotoxicity-induced neuronal death.


Subject(s)
Axons/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Glutamic Acid/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Animals , Brain/metabolism , Brain/pathology , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Embryo, Mammalian , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Green Fluorescent Proteins/genetics , Infarction, Middle Cerebral Artery/pathology , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mutation/genetics , Nerve Tissue Proteins/genetics , Neurons/drug effects , Phosphorylation/drug effects , Semaphorin-3A/pharmacology , Signal Transduction/drug effects , Transfection/methods , Tubulin/metabolism
12.
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
13.
Int Rev Cell Mol Biol ; 267: 125-81, 2008.
Article in English | MEDLINE | ID: mdl-18544498

ABSTRACT

Successful axonal outgrowth in the adult central nervous system (CNS) is central to the process of nerve regeneration and brain repair. To date, much of the knowledge on axonal guidance and outgrowth comes from studies on neuritogenesis and patterning during development where distal growth cones constantly sample the local environment and respond to specific physical and trophic influences. Opposing permissive (e.g., growth factors) and hostile signals (e.g., repulsive cues) are processed, leading to growth cone remodelling, and a concomitant restructuring of the cytoskeleton, thereby permitting pioneering extension and a potential for establishing synaptic connections. Repulsive cues, such as semaphorins, ephrins and myelin-secreted inhibitory glycoproteins, act through their respective receptors to affect the collapsing or turning of growth cones via several pathways, such as the Rho GTPases signalling which precipitates the cytoskeletal changes. One of the direct modulators of microtubules is the family of brain-specific proteins, collapsin response mediator protein (CRMP). Exciting evidence emerged recently that cleavage of CRMPs in response to injury-activated proteases, such as calpain, signals axonal retraction and neuronal death in adult post-mitotic neurons, while blocking this signal transduction prevents axonal retraction and death following excitotoxic insult and cerebral ischemia. Regeneration is minimal in injured postnatal CNS, albeit the occurrence of some limited remodelling in areas where synaptic plasticity is prevalent. Frequently in the absence of axonal regeneration, there is not only an inevitable loss of functional connections, but also a loss of neurons, such as through the actions of dependence receptors. Deciphering the cues and signalling pathways of axonal guidance and outgrowth may hold the key to fully understanding nerve regeneration and brain repair, thereby opening the way for developing potential therapeutics.


Subject(s)
Axons/physiology , Axons/ultrastructure , Cell Movement/physiology , Nerve Regeneration/physiology , Signal Transduction/physiology , Animals , Axons/pathology , Cell Polarity , Ephrin-A5/metabolism , Ephrins/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Glycoproteins/metabolism , Humans , Morphogenesis , Myelin Sheath/metabolism , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Netrin-1 , Neuronal Plasticity/physiology , Neuropilins/metabolism , Phosphoproteins/metabolism , Receptors, Immunologic/metabolism , Semaphorins/metabolism , Stroke/metabolism , Stroke/pathology , Stroke/therapy , Tumor Suppressor Proteins/metabolism , Roundabout Proteins
14.
Biochem Biophys Res Commun ; 367(1): 109-15, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18162177

ABSTRACT

Strategies to provide neuroprotection and to promote regenerative axonal outgrowth in the injured brain are thwarted by the plethora of axon growth inhibitors and the ligand promiscuity of some of their receptors. Especially, new neurons derived from ischemia-stimulated neurogenesis must integrate this multitude of inhibitory molecular cues, generated as a result of cortical damage, into a functional response. More often than not the response is one of growth cone collapse, axonal retraction and neuronal death. Therefore, characterization of the expression of inhibitory molecules in long-term surviving ischemic brains following stroke is important for designing selective therapeutics. Here, we describe a long-term recovery mouse model for cerebral ischemia in which a brief transient occlusion of the middle cerebral artery (30min) was followed by up to 30 days of long-term reperfusion. Significantly decreased grip strength motor function and increased expression of one of the major repulsive guidance cues, Semaphorin 3A (Sema3A) and its receptor Neuropilin1 (NRP1) occurred in brains of these mice. Interestingly, increased Doublecortin (DCX) expression occurred only in the lateral ventricular wall zone, but not in the dentate gyrus granule cell layer on the ischemic side of the brain. Importantly, no DCX positive cells were detected in the infarct core region after 30d ischemic recovery. Collectively, these studies demonstrated the sustained elevation of Sema3A/NRP1 expression in the ischemic territory, which may contribute to the inhibitory microenvironment responsible for preventing new neurons from entering the infarct area. This model will be of use as a platform for testing anti-inhibitory therapies to stroke.


Subject(s)
Brain Ischemia , Gene Expression Regulation/physiology , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Neuropilin-1/metabolism , Semaphorin-3A/metabolism , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/therapy , Doublecortin Domain Proteins , Doublecortin Protein , Gene Expression Regulation/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Models, Biological , Neuropeptides/genetics , Neuropilin-1/genetics , Semaphorin-3A/genetics , Stroke/metabolism , Stroke/pathology , Stroke/therapy , Time Factors , Treatment Outcome , Up-Regulation/genetics , Up-Regulation/physiology
15.
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
16.
Am J Pathol ; 170(5): 1725-38, 2007 May.
Article in English | MEDLINE | ID: mdl-17456777

ABSTRACT

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are both characterized pathologically by the presence of neuronal inclusions termed Lewy bodies (LBs). A common feature found in LBs are aggregates of alpha-synuclein (alpha-Syn), and although it is now recognized that alpha-Syn is the major building block for these toxic filaments, the mechanism of how this occurs remains unknown. In the present study, we demonstrate that proteolytic processing of alpha-Syn by the protease calpain I leads to the formation of aggregated high-molecular weight species and adoption of a beta-sheet structure. To determine whether calpain-cleavage of alpha-Syn occurs in PD and DLB, we designed site-directed calpain-cleavage antibodies to alpha-Syn and tested their utility in several animal model systems. Detection of calpain-cleaved alpha-Syn was evident in mouse models of cerebral ischemia and PD and in a Drosophila model of PD. In the human PD and DLB brain, calpain-cleaved alpha-Syn antibodies immunolabeled LBs and neurites in the substantia nigra. Moreover, calpain-cleaved alpha-Syn fragments identified within LBs colocalized with activated calpain in neurons of the PD and DLB brains. These findings suggest that calpain I may participate in the disease-linked aggregation of alpha-Syn in various alpha-synucleinopathies.


Subject(s)
Brain/metabolism , Calpain/metabolism , Lewy Body Disease/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Aged , Animals , Area Under Curve , Blotting, Western , Cell Line, Tumor , Drosophila , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunoprecipitation , Lewy Body Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neurons/metabolism , Parkinson Disease/pathology , Protein Structure, Secondary , alpha-Synuclein/chemistry
17.
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
18.
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
19.
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
20.
J Biol Chem ; 280(40): 33811-8, 2005 Oct 07.
Article in English | MEDLINE | ID: mdl-16091365

ABSTRACT

Minocycline is a potent neuroprotective tetracycline in animal models of cerebral ischemia. We examined the protective properties of chlortetracycline (CTC) and demeclocycline (DMC) and showed that these two tetracyclines were also potent neuroprotective against glutamate-induced neuronal death in vitro and cerebral ischemia in vivo. However, CTC and DMC appeared to confer neuroprotection through a unique mechanism compared with minocycline. Rather than inhibiting microglial activation and caspase, CTC and DMC suppressed calpain activities. In addition, CTC and DMC only weakly antagonized N-methyl-D-aspartate (NMDA) receptor activities causing 16 and 14%, respectively, inhibition of NMDA-induced whole cell currents and partially blocked NMDA-induced Ca2+ influx, commonly regarded as the major trigger of neuronal death. In vitro and in vivo experiments demonstrated that the two compounds selectively inhibited the activities of calpain I and II activated following glutamate treatment and cerebral ischemia. In contrast, minocycline did not significantly inhibit calpain activity. Taken together, these results suggested that CTC and DMC provide neuroprotection through suppression of a rise in intracellular Ca2+ and inhibition of calpains.


Subject(s)
Anti-Bacterial Agents/pharmacology , Brain Ischemia/prevention & control , Calpain/metabolism , Chlortetracycline/pharmacology , Demeclocycline/pharmacology , Glutamic Acid/toxicity , Neurons/drug effects , Animals , Brain Ischemia/physiopathology , Calcium/metabolism , Calpain/antagonists & inhibitors , Cell Culture Techniques , Enzyme Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...