Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Mol Neurosci ; 46(2): 303-14, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21691801

ABSTRACT

The presence of misfolded proteins elicits cellular responses including an endoplasmic reticulum (ER) stress response that may protect cells against the toxic buildup of misfolded proteins. Accumulation of these proteins in excessive amounts, however, overwhelms the "cellular quality control" system and impairs the protective mechanisms designed to promote correct folding and degrade misfolded proteins, ultimately leading to organelle dysfunction and cell death. Studies from multiple laboratories have identified the roles of several ER stress-induced cell death modulators and effectors. Earlier, we reported the role of the small co-chaperone protein p23 in preventing ER stress-induced cell death. p23 undergoes caspase-dependent cleavage to yield a 19-kD product (p19), and mutation of this caspase cleavage site not only blocks the formation of the 19-kD product but also attenuates the ER stress-induced cell death process triggered by various stressors. Thus, a critical question is whether p23 and/or p19 could serve as an in vivo marker for neurodegenerative diseases featuring misfolded proteins and cellular stress. In the present study, we used an antibody that recognizes both p23 and p19 as well as a specific neo-epitope antibody that detects only the p19 fragment. These antibodies were used to detect the presence of both these proteins in cells, primary neurons, brain samples from a mouse model of Alzheimer's disease (AD), and fixed human AD brain samples. While patients with severe AD did display a consistent reduction in p23 levels, our inability to observe p19 in mouse or human AD brain samples suggests that the usefulness of the p23 neo-epitope antibody is restricted to cells and primary neurons undergoing cellular stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Intramolecular Oxidoreductases/physiology , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Apoptosis/physiology , Brain/metabolism , Brain/pathology , Cell Hypoxia/physiology , Cells, Cultured/metabolism , Cytosol/chemistry , Disease Models, Animal , Epitopes/immunology , Female , Fibroblasts/metabolism , HEK293 Cells/metabolism , Humans , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/immunology , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Molecular Sequence Data , Neurons/metabolism , Prostaglandin-E Synthases , Recombinant Fusion Proteins/physiology , Thapsigargin/pharmacology , Transfection
2.
J Mol Neurosci ; 44(2): 91-102, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21249466

ABSTRACT

Previously, we identified valosin-containing protein (VCP) as a mediator of ER stress-induced cell death. Mutations in the VCP gene including R93, R155, and R191 have been described that manifest clinically as hereditary inclusion body myopathy with Paget's disease of bone and frontotemporal dementia. In addition, other studies have demonstrated that as a consequence of a mutation generated in the second ATP binding domain of VCP (K524A), cells accumulated large cytoplasmic vacuoles and underwent programmed cell death. In order to better understand the biochemical and molecular consequences of the clinically relevant VCP mutations as well as the genetically engineered ATPase-inactive mutant K524A and any relationship these may have to ER stress-induced cell death, we introduced analogous mutations separately and together into the human VCP gene and evaluated their effect on proteasome activity, Huntingtin protein aggregation and ER stress-induced cell death. Our results indicate that the VCP K524A mutant and the triple mutant VCP R93C-R155C-K524A block protein degradation, trigger Huntingtin aggregate formation, and render cells highly susceptible to ER stress-induced cell death as compared to VCPWT or other VCP mutants.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Mutation , Nerve Degeneration/pathology , Phenotype , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Cell Death/genetics , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Huntingtin Protein , Nerve Degeneration/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Thapsigargin/metabolism , Ubiquitin/metabolism , Valosin Containing Protein
3.
Ann Neurol ; 65(3): 294-303, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19334058

ABSTRACT

OBJECTIVE: Selective neuronal vulnerability in neurodegenerative diseases is poorly understood. In Alzheimer's disease, the basal forebrain cholinergic neurons are selectively vulnerable, putatively because of their expression of the cell death mediator p75(NTR) (the common neurotrophin receptor), and its interaction with proapoptotic ligands pro-nerve growth factor and amyloid-beta peptide. However, the relation between amyloid precursor protein (APP) and p75(NTR) has not been described previously. METHODS: APP and p75(NTR) were assayed for interaction by coimmunoprecipitation in vitro and in vivo, yeast two-hybrid assay, bioluminescence resonance energy transfer, and confocal microscopy. Effects on APP processing and signaling were studied using immunoblotting, enzyme-linked immunosorbent assays, and luciferase reporter assays. RESULTS: The results of this study are as follows: (1) p75(NTR) and APP interact directly; (2) this interaction is modified by ligands nerve growth factor and beta-amyloid; (3) APP and p75(NTR) colocalization in vivo is modified in Alzheimer's model transgenic mice; (4) APP processing is altered by p75(NTR), and to a lesser extent, p75(NTR) processing is altered by the presence of APP; (5) APP-dependent transcription mediated by Fe65 is blocked by p75(NTR); and (6) coexpression of APP and p75(NTR) triggers cell death. INTERPRETATION: These results provide new insight into the emerging signaling network that mediates the Alzheimer's phenotype and into the mechanism of basal forebrain cholinergic neuronal selective vulnerability. In addition, the results argue that the interaction between APP and p75(NTR) may represent a therapeutic target in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Death/genetics , Cell Line, Tumor , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Green Fluorescent Proteins/genetics , Humans , Immunoprecipitation/methods , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Neuroblastoma , Nuclear Proteins/metabolism , Protein Binding/drug effects , Rats , Receptors, Nerve Growth Factor/genetics , Transfection/methods , Two-Hybrid System Techniques
4.
Behav Brain Res ; 191(2): 246-55, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18485495

ABSTRACT

The deficits of Alzheimer's disease (AD) are believed to result, at least in part, from neurotoxicity of beta-amyloid (Abeta), a set of 38-43 amino acid fragments derived from the beta-amyloid precursor protein (APP). In addition, APP generates the APP-C31 and Jcasp toxic fragments intracellularly by cleavage at Asp664. We reported that mutation of Asp664 to Ala in a FAD-human APP transgene prevented AD-like deficits but did not affect Abeta production or deposition in PDAPP mice, arguing that D664A plays a crucial role in the generation of AD-like deficits. Whether D664A simply delays or completely prevents AD-like deficits, however, remained undefined. To address this question, we performed behavioral studies longitudinally on a pretrained mouse cohort at 9 and 13 months (mo) of age. While behavioral deficits were present in PDAPP mice, performance of Tg PDAPP(D664A) mice was not significantly different from non-Tg littermates' across all ages tested. Moreover, aberrant patterns in non-cognitive components of behavior in PDAPP mice were ameliorated in PDAPP(D664A) animals as well. A trend towards poorer retention at 9 mo and poorer learning at 13 mo that did not reach statistical significance was observed in PDAPP(D664A) mice. These results support and extend recent studies showing that cleavage of APP at Asp664 (or protein-protein interactions dependent on Asp664) is a crucial event in the generation of AD-like deficits in PDAPP mice. Our results thus further demonstrate that the D664A mutation either completely precludes, or markedly delays (beyond 13 mo) the appearance of AD-like deficits in this mouse model of AD.


Subject(s)
Alanine/genetics , Alzheimer Disease/complications , Amyloid beta-Protein Precursor/genetics , Aspartic Acid/genetics , Mental Disorders/etiology , Mutation/genetics , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Behavior, Animal , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Humans , Maze Learning , Mental Disorders/genetics , Mental Disorders/prevention & control , Mice , Mice, Transgenic , Reaction Time/genetics , Swimming
5.
J Alzheimers Dis ; 13(1): 1-16, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18334752

ABSTRACT

In addition to the proteolytic cleavages that give rise to amyloid-beta (Abeta), the amyloid-beta protein precursor (AbetaPP) is cleaved at Asp664 intracytoplasmically. This cleavage releases a cytotoxic peptide, APP-C31, removes AbetaPP-interaction motifs required for signaling and internalization, and is required for the generation of AD-like deficits in a mouse model of the disease. Although we and others had previously shown that Asp664 cleavage of AbetaPP is increased in AD brains, the distribution of the Asp664-cleaved forms of AbetaPP in non-diseased and AD brains at different ages had not been determined. Confirming previous reports, we found that Asp664-cleaved forms of AbetaPP were increased in neuronal cytoplasm and nuclei in early-stage AD brains but were absent in age-matched, non-diseased control brains and in late-stage AD brains. Remarkably, however, Asp664-cleaved AbetaPP was prominent in neuronal somata and in processes in entorhinal cortex and hippocampus of non-diseased human brains at ages <45 years. Our observations suggest that Asp664 cleavage of AbetaPP may be part of the normal proteolytic processing of AbetaPP in young (<45 years) human brain and that this cleavage is down-regulated with normal aging, but is aberrantly increased and altered in location in early AD.


Subject(s)
Alcohol Oxidoreductases/genetics , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Aspartic Acid/genetics , Cleavage Stimulation Factor/genetics , DNA-Binding Proteins/genetics , Genes, Switch/genetics , Aged , Aged, 80 and over , Alcohol Oxidoreductases/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid/metabolism , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Hippocampus/metabolism , Humans , Male , Signal Transduction/physiology
6.
FASEB J ; 22(6): 1737-47, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18198211

ABSTRACT

The signal transduction pathways involved in neuronal death are not well understood. Neuroglobin (Ngb), a recently discovered vertebrate globin expressed predominantly in the brain, shows increased expression in neurons in response to oxygen deprivation and protects neurons from ischemic and hypoxic death. The mechanism of this neuroprotection is unclear. We examined the surface distribution of raft membrane microdomains in cortical neuron cultures during hypoxia using the raft marker cholera toxin B (CTx-B) subunit. Mechanistically, we demonstrate that hypoxia induces rapid polarization of somal membranes and aggregation of microdomains with the subjacent mitochondrial network. This signaling complex is formed well before neurons commit to die, consistent with an early role in death signal transduction. Neurons from Ngb-overexpressing transgenic (Ngb-Tg) mice do not undergo microdomain polarization or mitochondrial aggregation in response to, and are resistant to death from hypoxia. We link the protective actions of Ngb to inhibition of Pak1 kinase activity and Rac1-GDP-dissociation inhibitor disassociation, and inhibition of actin assembly and death-signaling module polarization.


Subject(s)
Globins/physiology , Hypoxia/metabolism , Nerve Tissue Proteins/physiology , Neurons/pathology , Signal Transduction , Actins/antagonists & inhibitors , Animals , Cerebral Cortex , Membrane Microdomains/metabolism , Membrane Microdomains/ultrastructure , Mice , Mice, Transgenic , Neuroglobin , p21-Activated Kinases/antagonists & inhibitors
7.
J Neurochem ; 104(4): 1065-80, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17986220

ABSTRACT

The deficits in Alzheimer disease (AD) stem at least partly from neurotoxic beta-amyloid peptides generated from the amyloid precursor protein (APP). APP may also be cleaved intracellularly at Asp664 to yield a second neurotoxic peptide, C31. Previously, we showed that cleavage of APP at the C-terminus is required for the impairments seen in APP transgenic mice, by comparing elements of the disease in animals modeling AD, with (platelet-derived growth factor B-chain promoter-driven APP transgenic mice; PDAPP) versus without (PDAPP D664A) a functional Asp664 caspase cleavage site. However, the signaling mechanism(s) by which Asp664 contributes to these deficits remains to be elucidated. In this study, we identify a kinase protein, recently shown to bind APP at the C-terminus and to contribute to AD, whose activity is modified in PDAPP mice, but normalized in PDAPP D664A mice. Specifically, we observed a significant increase in nuclear p21-activated kinase (isoforms 1, 2, and or 3; PAK-1/2/3) activation in hippocampus of 3 month old PDAPP mice compared with non-transgenic littermates, an effect completely prevented in PDAPP D664A mice. In contrast, 13 month old PDAPP mice displayed a significant decrease in PAK-1/2/3 activity, which was once again absent in PDAPP D664A mice. Similarly, in hippocampus of early and severe AD subjects, there was a progressive and subcellular-specific reduction in active PAK-1/2/3 compared with normal controls. Interestingly, total PAK-1/2/3 protein was increased in early AD subjects, but declined in moderate AD and declined further, to significantly below that of control levels, in severe AD. These findings are compatible with previous suggestions that PAK may be involved in the pathophysiology of AD, and demonstrate that both early activation and late inactivation in the murine AD model require the cleavage of APP at Asp664.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid/metabolism , Peptide Fragments/physiology , Signal Transduction/physiology , p21-Activated Kinases/physiology , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid/genetics , Humans , Hydrolysis , Isoenzymes/genetics , Isoenzymes/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/genetics , Point Mutation/physiology , p21-Activated Kinases/chemistry , p21-Activated Kinases/genetics
8.
Proc Natl Acad Sci U S A ; 104(48): 19114-9, 2007 Nov 27.
Article in English | MEDLINE | ID: mdl-18025470

ABSTRACT

Neuroglobin (Ngb), a vertebrate globin expressed primarily in neurons, is induced by and protects against neuronal hypoxia and cerebral ischemia. To investigate the spectrum and mechanism of Ngb's neuroprotective action, we studied the effect of transgenic overexpression of Ngb on NMDA and beta-amyloid (Abeta) toxicity in murine cortical neuron cultures in vitro and on the phenotype of Alzheimer's disease (AD) transgenic (APP(Sw,Ind)) mice. Compared with cortical neuron cultures from wild-type mice, cultures from Ngb-overexpressing transgenic (Ngb-Tg mice) were resistant to the toxic effects of NMDA and Abeta(25-35), as measured by polarization of cell membrane lipid rafts, mitochondrial aggregation, lactate dehydrogenase release, and nuclear fragmentation. In addition, compared with APP(Sw,Ind) mice, double-transgenic (Ngb-Tg x APP(Sw,Ind)) mice showed reductions in thioflavin-S-stained extracellular Abeta deposits, decreased levels of Abeta(1-40) and Abeta(1-42), and improved behavioral performance in a Y-maze test of spontaneous alternations. These findings suggest that the spectrum of Ngb's neuroprotective action extends beyond hypoxic-ischemic insults. Ngb may protect neurons from NMDA and Abeta toxicity by inhibiting the formation of a death-signaling membrane complex, and interventions that increase Ngb expression could have therapeutic application in AD and other neurodegenerative disorders.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Globins/physiology , Membrane Microdomains/pathology , Nerve Tissue Proteins/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/analysis , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/toxicity , Animals , Brain Chemistry , Cells, Cultured/pathology , Crosses, Genetic , Globins/genetics , Humans , Maze Learning , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Neuroglobin , Neurons/pathology , Peptide Fragments/analysis , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology , Recombinant Fusion Proteins/toxicity , Single-Blind Method
9.
Neurobiol Dis ; 28(1): 65-75, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17719230

ABSTRACT

The amyloid precursor protein (APP) is a type I transmembrane protein translocated to neuronal terminals, whose function is still unknown. The C-terminus of APP mediates its interaction with cellular adaptor and signaling proteins, some of which signal to the stress-activated protein kinase (SAPK) pathway. Here we show that ASK1, a MAPKKK that activates two SAPKs, c-Jun N-terminal-kinase (JNK) and p38, is present in a complex containing APP, phospho-MKK6, JIP1 and JNK1. In primary neurons deprived of growth factors, as well as in brains of (FAD)APP-transgenic mice, ASK1 was upregulated in neuronal projections, where it interacted with APP. In non-transgenic brains, ASK1 and APP associated mainly in the ER. Our results indicate that recruitment of ASK1 to stress-signaling complexes assembled with APP may be triggered and enhanced by cellular stress. Thus, ASK1 may be the apical MAPKKK in a signaling complex assembled with APP as a response to stress.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Amyloid beta-Protein Precursor/chemistry , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , MAP Kinase Kinase 6/metabolism , MAP Kinase Kinase Kinase 5/chemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Mitogen-Activated Protein Kinase 8/metabolism , Neurons/chemistry , Protein Transport/physiology , Synaptic Vesicles/metabolism
10.
Mol Pharmacol ; 71(6): 1525-34, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17392524

ABSTRACT

A major challenge to broadening oncology applications for inhibitors of the ubiquitin-proteasome system (UPS) is the identification of UPS-dependent cancer pathways predictive of tumors responsive to peptidomimetic inhibitors of its 20S core protease activity. To inform clinical studies evaluating UPS inhibitors as breast cancer therapeutics, seven phenotypically diverse human breast cancer cell line models were characterized for their cellular and molecular responses to the clinically approved 20S inhibitor bortezomib (PS341; Velcade), focusing on those overexpressing estrogen receptor (ER) or ERBB2/HER2, because these oncogenic receptor pathways are constitutively activated in approximately 80% of all breast cancers. All models demonstrated dose-dependent bortezomib reduction in intracellular 20S activity correlating with cell growth inhibition, and bortezomib IC(50) values (concentrations producing 50% growth inhibition) varied directly with pretreatment 20S activities (r = 0.74; *, p < 0.05), suggesting that basal 20S activity may serve as a clinical predictor of tumor responsiveness to UPS inhibition. Reduction in 20S activity (> 60%) was associated with early (24 h) intracellular relocalization of ER (nucleus to cytoplasm) and ERBB2 (plasma membrane to perinuclear lysosomes), buildup of ubiquitinated and Hsp70-associated receptor, degradation and loss of ER and ERBB2 function, and induction of cellular apoptosis. These models were also used to screen a pharmacologic panel of pathway-targeted anticancer agents [4-hydroxy-3-methoxy-5-(benzothiazolylthiomethyl)benzylidenecyanoacetamide (AG825), 6-(4-bromo-2-chloro-phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxy-ethoxy)-amide (AZD6244/ARRY142886), 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one hydrochloride (LY294002), 17-N-allylamino-17-demethoxy geldanamycin (17AAG), and (2E)-N-hydroxy-3-[4-[[(2-hydroxyethyl)[2-(1H-indol-3-yl)ethyl]amino]methyl]phenyl]-2-propenamide (LAQ824)] for those capable of sensitizing to bortezomib. In keeping with the observation that 20S reduction has little effect on mitogen-activated protein kinase kinase 1/2 (MEK1/2) signaling in either ER-positive or ERBB2-positive models, only the MEK-1/2 inhibitor AZD6244 consistently improved the antitumor activity of bortezomib.


Subject(s)
Breast Neoplasms/metabolism , Proteasome Endopeptidase Complex/physiology , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Breast Neoplasms/drug therapy , Drug Therapy, Combination , Humans , Oxidation-Reduction , Proteasome Inhibitors , Pyrazines/pharmacology , Transcription, Genetic/drug effects
11.
J Neurosci ; 27(2): 304-7, 2007 Jan 10.
Article in English | MEDLINE | ID: mdl-17215390

ABSTRACT

We sought genetic evidence for the involvement of neuronal vascular endothelial growth factor (VEGF) in amyotrophic lateral sclerosis (ALS). Mice expressing human ALS mutant superoxide dismutase-1 (SOD1) were crossed with mice that overexpress VEGF in neurons (VEGF+/+). We report that SOD1(G93A)/VEGF+/+ double-transgenic mice show delayed motor neuron loss, delayed motor impairment, and prolonged survival compared with SOD1(G93A) single transgenics. These findings indicate that neuronal VEGF protects against motor neuron degeneration, and may have therapeutic implications for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Gene Expression Regulation/physiology , Nerve Degeneration/metabolism , Nerve Degeneration/prevention & control , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Amyotrophic Lateral Sclerosis/mortality , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Gene Expression Regulation, Enzymologic/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/pathology , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival Rate , Vascular Endothelial Growth Factor A/physiology
12.
J Neurosci Res ; 85(4): 740-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17243175

ABSTRACT

New neurons are generated continuously in the subventricular zone and dentate gyrus of the adult brain. Neuropathologic processes, including cerebral ischemia, can enhance neurogenesis, as can growth factors and other physiologic stimuli. Vascular endothelial growth factor (VEGF) is an angiogenic and neuroprotective growth factor that can promote neurogenesis, but it is unknown whether VEGF can enhance migration of newborn neurons toward sites of ischemic injury, where they might be able to replace neurons that undergo ischemic death. In the present study we produced permanent focal cerebral ischemia in transgenic (Tg) mice that overexpress VEGF. Cell proliferation and neurogenesis were assessed with bromodeoxyuridine (Brdu) labeling and immunostaining for cell type-specific markers. In VEGF-Tg mice, brains examined 7-28 days after cerebral ischemia showed markedly increased subventricular zone (SVZ) neurogenesis, chains of neuroblasts extending from the SVZ to the peri-infarct cortex, and an increase in the number of newly generated cortical neurons at 14-28 days after ischemia. In concert with these effects, VEGF overexpression reduced infarct volume and improved postischemic motor function. These findings provide evidence that VEGF increases SVZ neurogenesis and neuromigration, consistent with a possible role in repair. Our data suggest that in addition to its neuroprotective effects, which are associated with improved outcome in the acute phase after cerebral ischemia, VEGF enhances postischemic neurogenesis, which could provide a therapeutic target for more chronic brain repair.


Subject(s)
Cell Movement/genetics , Cell Proliferation , Gene Expression Regulation/physiology , Infarction, Middle Cerebral Artery/pathology , Neurons/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Brain Infarction/etiology , Brain Infarction/pathology , Bromodeoxyuridine/metabolism , Cell Count/methods , Disease Models, Animal , Functional Laterality , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Neurons/pathology , Phosphopyruvate Hydratase/metabolism , Time Factors
13.
Brain Res ; 1123(1): 237-44, 2006 Dec 06.
Article in English | MEDLINE | ID: mdl-17064673

ABSTRACT

Basic fibroblast growth factor (FGF-2) has been reported to protect against ischemic injury in the brains of young adult rodents. However, little is known about whether FGF-2 retains this capability in the aged ischemic brain. Since stroke in human is much more common in older people than among younger adults, to address this question is clinically important. In this study, aged (24-month-old) rats were treated with intracerebroventricular infusion of FGF-2 or vehicle for 3 days, beginning 48 h before (pre-ischemia), 24 h after (early post-ischemia), or 96 h after (late post-ischemia) 60 min of middle cerebral artery occlusion, and were killed 10 days after ischemia. Aged rats given FGF-2 pre-ischemia showed better symmetry of movement and forepaw outstretching, and reduced infarct volumes, compared to rats treated with vehicle, but no significant improvement was found in aged rats given FGF-2 after focal ischemia. In contrast, young adult (3-month-old) rats treated with FGF-2 for 3 days beginning 24 h post-ischemia showed significant neurobehavioral improvement and better histological outcome. In addition, we also found that newborn neurons in the rostral subventricular zone (SVZ) were increased in aged rats treated with FGF-2 prior to ischemia. However, unlike in young adult ischemic rats, only a few of newly generated cells migrated into the damaged region in aged brain after focal ischemia. These findings point to differences in the response of aged versus young adult rats to FGF-2 in cerebral ischemia, and suggest that such differences need to be considered in the development of neuroprotective agents for stroke.


Subject(s)
Brain Infarction/prevention & control , Fibroblast Growth Factor 2/metabolism , Nerve Regeneration/physiology , Recovery of Function/drug effects , Stroke/drug therapy , Age Factors , Analysis of Variance , Animals , Brain Infarction/drug therapy , Disease Models, Animal , Drug Administration Schedule , Fibroblast Growth Factor 2/administration & dosage , Injections, Intraventricular , Male , Nerve Regeneration/drug effects , Neuroprotective Agents/administration & dosage , Rats , Rats, Inbred F344
14.
Proc Natl Acad Sci U S A ; 103(35): 13198-202, 2006 Aug 29.
Article in English | MEDLINE | ID: mdl-16924107

ABSTRACT

Experimental stroke in rodents stimulates neurogenesis and migration of newborn neurons from their sites of origin into ischemic brain regions. We report that in patients with stroke, cells that express markers associated with newborn neurons are present in the ischemic penumbra surrounding cerebral cortical infarcts, where these cells are preferentially localized in the vicinity of blood vessels. These findings suggest that stroke-induced compensatory neurogenesis may occur in the human brain, where it could contribute to postischemic recovery and represent a target for stroke therapy.


Subject(s)
Cell Differentiation , Cerebral Cortex/cytology , Neurons/cytology , Stroke/pathology , Stroke/physiopathology , Adult , Aged , Brain Ischemia , Cell Proliferation , Cerebral Cortex/pathology , Female , Humans , Male , Middle Aged
15.
Proc Natl Acad Sci U S A ; 103(18): 7130-5, 2006 May 02.
Article in English | MEDLINE | ID: mdl-16641106

ABSTRACT

The deficits characteristic of Alzheimer's disease (AD) are believed to result, at least in part, from the neurotoxic effects of beta-amyloid peptides, a set of 39-43 amino acid fragments derived proteolytically from beta-amyloid precursor protein (APP). APP also is cleaved intracytoplasmically at Asp-664 to generate a second cytotoxic peptide, APP-C31, but whether this C-terminal processing of APP plays a role in the pathogenesis of AD is unknown. Therefore, we compared elements of the Alzheimer's phenotype in transgenic mice modeling AD with vs. without a functional Asp-664 caspase cleavage site. Surprisingly, whereas beta-amyloid production and plaque formation were unaltered, synaptic loss, astrogliosis, dentate gyral atrophy, increased neuronal precursor proliferation, and behavioral abnormalities were completely prevented by a mutation at Asp-664. These results suggest that Asp-664 plays a critical role in the generation of Alzheimer-related pathophysiological and behavioral changes in human APP transgenic mice, possibly as a cleavage site or via protein-protein interactions.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Aspartic Acid/metabolism , Behavior, Animal/physiology , Point Mutation , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Proliferation , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Maze Learning/physiology , Mice , Mice, Transgenic , Neurons/cytology , Neurons/physiology , Stem Cells/cytology , Stem Cells/physiology
16.
Ann Neurol ; 58(2): 277-89, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16049941

ABSTRACT

Several approaches have been used in an effort to identify proteins that interact with beta-amyloid precursor protein (APP). However, few studies have addressed the identification of proteins associated with APP in brain tissue from patients with Alzheimer's disease. We report the results of a pilot proteomic study performed on complexes immunoprecipitated with APP in brain samples of patients with Alzheimer's disease and normal control subjects. The 21 proteins identified could be grouped into five functional classes: molecular chaperones, cytoskeletal and structural proteins, proteins involved in trafficking, adaptors, and enzymes. Among the proteins identified, six had been reported previously as direct, indirect, or genetically inferred APP interactors. The other 15 proteins immunoprecipitated with APP were novel potential partners. We confirmed the APP interaction by Western blotting and coimmunolocalization in brain tissues, for 5 of the 21 interactors. In agreement with previous studies, our results are compatible with an involvement of APP in axonal transport and vesicular trafficking, and with a potential association of APP with cellular protein folding/protein degradation systems.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Nerve Tissue Proteins/metabolism , Proteomics/methods , Amyloid beta-Protein Precursor/chemistry , Blotting, Western/methods , Brain/pathology , Crystallins/metabolism , Dynamins/metabolism , Electrophoresis, Gel, Two-Dimensional/methods , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Pilot Projects
17.
Neurobiol Dis ; 18(2): 366-74, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15686965

ABSTRACT

Cell replacement therapy may have the potential to promote brain repair and recovery after stroke. To compare how focal cerebral ischemia affects the entry, migration, and phenotypic features of neural precursor cells transplanted by different routes, we administered neuronal precursors from embryonic cerebral cortex of green fluorescent protein (GFP)-expressing transgenic mice to rats that had undergone middle cerebral artery occlusion (MCAO) by the intrastriatal, intraventricular, and intravenous routes. MCAO increased the entry of GFP-immunoreactive cells, most of which expressed neuroepithelial (nestin) or neuronal (doublecortin) markers, from the ventricles and bloodstream into the brain, and enhanced their migration when delivered by any of these routes. Transplanted neural precursors migrated into the ischemic striatum and cerebral cortex. Thus, transplantation of neural precursors by a variety of routes can deliver cells with the potential to replace injured neurons to ischemic brain regions.


Subject(s)
Brain Ischemia/therapy , Cell Movement/physiology , Stem Cell Transplantation/methods , Stem Cells/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Corpus Striatum/cytology , Corpus Striatum/physiology , Corpus Striatum/surgery , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Graft Survival/physiology , Infarction, Middle Cerebral Artery/therapy , Infusions, Intravenous , Injections, Intraventricular , Intermediate Filament Proteins/metabolism , Male , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Neuropeptides/metabolism , Phenotype , Rats , Rats, Sprague-Dawley , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...