Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Proc Natl Acad Sci U S A ; 106(29): 12145-50, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19581601

ABSTRACT

A number of distinct beta-amyloid (Abeta) variants or multimers have been implicated in Alzheimer's disease (AD), and antibodies recognizing such peptides are in clinical trials. Humans have natural Abeta-specific antibodies, but their diversity, abundance, and function in the general population remain largely unknown. Here, we demonstrate with peptide microarrays the presence of natural antibodies against known toxic Abeta and amyloidogenic non-Abeta species in plasma samples and cerebrospinal fluid of AD patients and healthy controls aged 21-89 years. Antibody reactivity was most prominent against oligomeric assemblies of Abeta and pyroglutamate or oxidized residues, and IgGs specific for oligomeric preparations of Abeta1-42 in particular declined with age and advancing AD. Most individuals showed unexpected antibody reactivities against peptides unique to autosomal dominant forms of dementia (mutant Abeta, ABri, ADan) and IgGs isolated from plasma of AD patients or healthy controls protected primary neurons from Abeta toxicity. Aged vervets showed similar patterns of plasma IgG antibodies against amyloid peptides, and after immunization with Abeta the monkeys developed high titers not only against Abeta peptides but also against ABri and ADan peptides. Our findings support the concept of conformation-specific, cross-reactive antibodies that may protect against amyloidogenic toxic peptides. If a therapeutic benefit of Abeta antibodies can be confirmed in AD patients, stimulating the production of such neuroprotective antibodies or passively administering them to the elderly population may provide a preventive measure toward AD.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Antibodies/immunology , Neuroprotective Agents/immunology , Peptides/immunology , Aging/drug effects , Alzheimer Disease/blood , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Antibodies/blood , Antibodies/cerebrospinal fluid , Cytoprotection/drug effects , Dementia/complications , Dementia/immunology , Disease Progression , Genes, Dominant , Immunization , Immunoglobulin G/blood , Mice , Molecular Weight , Neurons/cytology , Neurons/drug effects , Peptides/chemistry , Primates/immunology , Protein Processing, Post-Translational/drug effects , Protein Structure, Quaternary
2.
Nat Med ; 7(3): 338-43, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231633

ABSTRACT

Clusterin, also known as apolipoprotein J, is a ubiquitously expressed molecule thought to influence a variety of processes including cell death. In the brain, it accumulates in dying neurons following seizures and hypoxic-ischemic (H-I) injury. Despite this, in vivo evidence that clusterin directly influences cell death is lacking. Following neonatal H-I brain injury in mice (a model of cerebral palsy), there was evidence of apoptotic changes (neuronal caspase-3 activation), as well as accumulation of clusterin in dying neurons. Clusterin-deficient mice had 50% less brain injury following neonatal H-I. Surprisingly, the absence of clusterin had no effect on caspase-3 activation, and clusterin accumulation and caspase-3 activation did not colocalize to the same cells. Studies with cultured cortical neurons demonstrated that exogenous purified astrocyte-secreted clusterin exacerbated oxygen/glucose-deprivation-induced necrotic death. These results indicate that clusterin may be a new therapeutic target to modulate non-caspase-dependent neuronal death following acute brain injury.


Subject(s)
Brain/pathology , Caspases/metabolism , Glycoproteins/physiology , Hypoxia-Ischemia, Brain/pathology , Molecular Chaperones/physiology , Animals , Animals, Newborn , Blotting, Western , Caspase 3 , Cell Death/physiology , Clusterin , Fluorescent Antibody Technique , Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Molecular Chaperones/genetics
3.
Brain Behav Immun ; 24(5): 759-67, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19945527

ABSTRACT

Cerebral periventricular white matter injury stands as a leading cause of cognitive, behavioral and motor impairment in preterm infants. There is epidemiological and histopathological evidence demonstrating the role of prenatal or neonatal inflammation in brain injury in preterm infants. In order to define the effect of an inflammatory insult in the developing brain on magnetic resonance (MR) imaging, we obtained high resolution conventional and diffusion MR images of the brain of rat pups after an inflammatory injury. Rat pups were subjected on postnatal day 5 (P5) to a stereotaxic injection of lipopolysaccharide in the corpus callosum and then imaged at 11.7 T on days 0, 2 and 4 following the injury. They were subsequently sacrificed for immunohistochemistry. Diffusion tensor imaging (DTI) acquired at high spatial resolution showed an initial reduction of the apparent diffusion coefficient (ADC) in the white matter. This was followed by an increase in ADC value and in T2 relaxation time constant in the white matter, with an associated increase of radial diffusivity of the corpus callosum, and a 10-fold increase in ventricular size. On histology, these MR changes corresponded to widespread astrogliosis, and decreased proportion of the section areas containing cresyl violet positive stain. The increase in radial diffusivity, typically attributed to myelin loss, occurred in this case despite the absence of myelin at this developmental stage.


Subject(s)
Brain Injuries/pathology , Brain/pathology , Animals , Animals, Newborn , Anisotropy , Image Processing, Computer-Assisted , Immunohistochemistry , Inflammation/pathology , Magnetic Resonance Imaging , Male , Nerve Fibers, Myelinated/pathology , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric
4.
Trends Biochem Sci ; 16(4): 140-4, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1877089

ABSTRACT

In the past decade, there has been an explosion of information relating to the molecular neurobiology of Alzheimer's disease (AD). Molecular dissection of the neuropathology of AD has provided insight into the pathogenesis of this disease and has defined areas where investigation may prove useful in elucidating the cause of this disorder and suggest new treatments.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Disease Models, Animal , Humans , Male
5.
Neuron ; 9(3): 465-78, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1524827

ABSTRACT

Nerve growth factor (NGF) appears to act as a neurotrophic factor for basal forebrain and caudate-putamen cholinergic neurons. The mechanism by which NGF transduces its signal in these neurons is yet to be defined. Recent data indicate that the product of the trk gene, p140trk, is a critical component of the NGF receptor. Herein, we show that p140trk mRNA is highly restricted in its distribution in the adult rat forebrain, that it is present in cholinergic neurons, and that most if not all cholinergic neurons contain p140trk mRNA. Furthermore, induction of trk expression by NGF suggests that neurotrophin-mediated up-regulation of their receptor tyrosine kinases is an important feature of their actions and that neurotrophins may regulate the activity of responsive neurons through increasing the level of their receptors.


Subject(s)
Gene Expression Regulation , Nerve Growth Factors/physiology , Neurons/physiology , Prosencephalon/physiology , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Animals , Caudate Nucleus/metabolism , Choline O-Acetyltransferase/genetics , Gene Expression Regulation/drug effects , Nerve Growth Factors/pharmacology , Neurons/metabolism , Putamen/metabolism , Receptor, trkA , Tissue Distribution
7.
J Clin Invest ; 103(6): R15-R21, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10079115

ABSTRACT

The epsilon4 allele of apolipoprotein E (apo E) is associated with an increased risk for developing Alzheimer's disease (AD). This may be due to interactions between apo E and the amyloid-beta protein (Abeta). To assess the effects of human apo E isoforms on Abeta deposition in vivo, we bred apo E3 and apo E4 hemizygous (+/-) transgenic mice expressing apo E by astrocytes to mice homozygous (+/+) for a mutant amyloid precursor protein (APPV717F) transgene that develop age-dependent AD neuropathology. All mice were on a mouse apo E null (-/-) background. By nine months of age, APPV717F+/-, apo E-/- mice had developed Abeta deposition, and, as reported previously, the quantity of Abeta deposits was significantly less than that seen in APPV717F+/- mice expressing mouse apo E. In contrast to effects of mouse apo E, similar levels of human apo E3 and apo E4 markedly suppressed early Abeta deposition at nine months of age in APPV717F+/- transgenic mice, even when compared with mice lacking apo E. These findings suggest that human apo E isoforms decrease Abeta aggregation or increase Abeta clearance relative to an environment in which mouse apo E or no apo E is present. The results may have important implications for understanding mechanisms underlying the link between apo E and AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoproteins E/biosynthesis , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Animals , Apolipoproteins E/genetics , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mutation , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Recombinant Proteins/biosynthesis
8.
J Clin Invest ; 101(9): 1992-9, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9576764

ABSTRACT

Programmed cell death (apoptosis) is a normal process in the developing nervous system. Recent data suggest that certain features seen in the process of programmed cell death may be favored in the developing versus the adult brain in response to different brain injuries. In a well characterized model of neonatal hypoxia-ischemia, we demonstrate marked but delayed cell death in which there is prominent DNA laddering, TUNEL-labeling, and nuclei with condensed chromatin. Caspase activation, which is required in many cases of apoptotic cell death, also followed a delayed time course after hypoxia-ischemia. Administration of boc-aspartyl(OMe)-fluoromethylketone, a pan-caspase inhibitor, was significantly neuroprotective when given by intracerebroventricular injection 3 h after cerebral hypoxia-ischemia. In addition, systemic injections of boc-aspartyl(OMe)-fluoromethylketone also given in a delayed fashion, resulted in significant neuroprotection. These findings suggest that caspase inhibitors may be able to provide benefit over a prolonged therapeutic window after hypoxic-ischemic events in the developing brain, a major contributor to static encephalopathy and cerebral palsy.


Subject(s)
Amino Acid Chloromethyl Ketones/therapeutic use , Brain Ischemia/drug therapy , Cysteine Proteinase Inhibitors/therapeutic use , Hypoxia/drug therapy , Amino Acid Chloromethyl Ketones/administration & dosage , Animals , Animals, Newborn , Apoptosis/drug effects , Carotid Arteries/surgery , Coumarins/metabolism , Cysteine Proteinase Inhibitors/administration & dosage , DNA Damage/drug effects , Injections, Intraperitoneal , Injections, Intraventricular , Ligation , Oligopeptides/metabolism , Rats , Rats, Sprague-Dawley , Staining and Labeling/methods , Time Factors
9.
J Prev Alzheimers Dis ; 4(4): 236-241, 2017.
Article in English | MEDLINE | ID: mdl-29181488

ABSTRACT

Tau neurofibrillary tangles are found in the brains of patients suffering from Alzheimer's disease and other tauopathies. The progressive spreading of tau pathology from one brain region to the next is believed to be caused by extracellular transsynaptic transmission of misfolded tau between neurons. Preclinical studies have shown that antibodies against tau can prevent this transfer of misfolded tau between cells. Thus, antibodies against tau have the potential to stop or slow the progression of tau pathology observed in human tauopathies. To test this hypothesis, a humanized anti-tau antibody (ABBV-8E12) was developed and a phase 1 clinical trial of this antibody has been completed. The double-blind, placebo-controlled phase 1 study tested single doses of ABBV-8E12 ranging from 2.5 to 50 mg/kg in 30 patients with progressive supranuclear palsy (PSP). ABBV-8E12 was found to have an acceptable safety profile with no clinically concerning trends in the number or severity of adverse events between the placebo and dosed groups. Pharmacokinetic modelling showed that the antibody has a plasma half-life and cerebrospinal fluid:plasma ratio consistent with other humanized antibodies, and there were no signs of immunogenicity against ABBV-8E12. Based on the acceptable safety and tolerability profile of single doses of ABBV-8E12, AbbVie is currently enrolling patients into two phase 2 clinical trials to assess efficacy and safety of multiple doses of ABBV-8E12 in patients with early Alzheimer's disease or PSP.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunologic Factors/therapeutic use , Tauopathies/therapy , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Double-Blind Method , Drug Evaluation, Preclinical , Humans , Immunologic Factors/adverse effects , Immunologic Factors/pharmacokinetics , Immunotherapy , Models, Biological , Tauopathies/blood , Tauopathies/cerebrospinal fluid
11.
J Neurosci ; 20(2): 542-9, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10632583

ABSTRACT

The low-density lipoprotein (LDL) receptor-related protein (LRP) is a multifunctional endocytic receptor that is expressed abundantly in neurons of the CNS. Both LRP and several of its ligands, including tissue plasminogen activator (tPA), apolipoprotein E/lipoproteins, alpha(2)-macroglobulin, and the beta-amyloid precursor protein, have been implicated in various neuronal functions and in the pathogenesis of Alzheimer's disease. It has been reported that induction of tPA expression may contribute to activity-dependent synaptic plasticity in the hippocampus and cerebellum. In addition, long-term potentiation (LTP) is significantly decreased in mice lacking tPA. Here we demonstrate that tPA receptor LRP is abundantly expressed in hippocampal neurons and participates in hippocampal LTP. Perfusion of hippocampal slices with receptor-associated protein (RAP), an antagonist for ligand interactions with LRP, significantly reduced late-phase LTP (L-LTP). In addition, RAP also blocked the enhancing effect of synaptic potentiation by exogenous tPA in hippocampal slices prepared from tPA knock-out mice. Metabolic labeling and ligand binding analyses showed that both tPA and LRP are synthesized by hippocampal neurons and that LRP is the major cell surface receptor that binds tPA. Finally, we found that tPA binding to LRP in hippocampal neurons enhances the activity of cyclic AMP-dependent protein kinase, a key molecule that is known to be involved in L-LTP. Taken together, our results demonstrate that interactions between tPA and cell surface LRP are important for hippocampal L-LTP.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Neurons/physiology , Receptors, Immunologic/physiology , Tissue Plasminogen Activator/metabolism , Animals , Carrier Proteins/pharmacology , Carrier Proteins/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Endocytosis , Glycoproteins/pharmacology , Glycoproteins/physiology , Humans , In Vitro Techniques , LDL-Receptor Related Protein-Associated Protein , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Neurons/cytology , Receptors, Immunologic/genetics , Recombinant Proteins/pharmacology , Tissue Plasminogen Activator/deficiency , Tissue Plasminogen Activator/genetics , alpha-Macroglobulins/physiology
12.
Cell Death Differ ; 10(10): 1148-55, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14502238

ABSTRACT

A single episode of ethanol intoxication triggers widespread apoptotic neurodegeneration in the infant rat or mouse brain. The cell death process occurs over a 6-16 h period following ethanol administration, is accompanied by a robust display of caspase-3 enzyme activation, and meets ultrastructural criteria for apoptosis. Two apoptotic pathways (intrinsic and extrinsic) have been described, either of which may culminate in the activation of caspase-3. The intrinsic pathway is regulated by Bax and Bcl-XL and involves Bax-induced mitochondrial dysfunction and release of cytochrome c as antecedent events leading to caspase-3 activation. Activation of caspase-8 is a key event preceding caspase-3 activation in the extrinsic pathway. In the present study, following ethanol administration to infant mice, we found no change in activated caspase-8, which suggests that the extrinsic pathway is not involved in ethanol-induced apoptosis. We also found that ethanol triggers robust caspase-3 activation and apoptotic neurodegeneration in C57BL/6 wildtype mice, but induces neither phenomenon in homozygous Bax-deficient mice. Therefore, it appears that ethanol-induced neuroapoptosis is an intrinsic pathway-mediated phenomenon involving Bax-induced disruption of mitochondrial membranes and cytochrome c release as early events leading to caspase-3 activation.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Ethanol/pharmacology , Neurons/drug effects , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Animals , Anterior Thalamic Nuclei/drug effects , Anterior Thalamic Nuclei/pathology , Blotting, Western , Brain/pathology , Brain Chemistry/drug effects , Caspase 3 , Caspase 8 , Caspases/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cytochromes c/analysis , Ethanol/blood , Genotype , Heterozygote , Hippocampus/drug effects , Hippocampus/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neurons/pathology , Protein Transport/drug effects , Protein Transport/physiology , Proto-Oncogene Proteins/genetics , Spectrin/analysis , Time Factors , Up-Regulation , bcl-2-Associated X Protein
13.
Trends Cardiovasc Med ; 8(6): 250-5, 1998 Aug.
Article in English | MEDLINE | ID: mdl-14987560

ABSTRACT

Apolipoprotein E (apoE) is a well characterized 299 amino acid protein that participates in the regulation of plasma cholesterol and lipid metabolism. In humans, apoE has three major protein isoforms: E2 (cys(112), cys(158)); E3 (cys(112), arg(158)); and E4 (arg(112), arg(158)) that are encoded for by a single gene on chromosome 19. Genetic studies have shown that apoE4 is a risk factor for Alzheimer's disease (AD) as well as for poor outcome following certain injuries to the central nervous system (CNS). These genetic data, as well as other data reviewed herein, suggest that apoE may play an important role in the nervous system under certain conditions. This review focuses on studies demonstrating that apoE can modulate neuronal structure and the potential implication of these findings for its role following CNS injury, in AD, and in other neurodegenerative diseases.

14.
Clin Pharmacol Ther ; 98(5): 469-71, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26250900

ABSTRACT

Alzheimer disease (AD) is the most common cause of dementia and is characterized by the aggregation and accumulation of two proteins in the brain, amyloid-ß (Aß) and tau. Aß and tau begin to buildup 15-20 years before the clinical onset of AD dementia. Increasing evidence suggests that preventing or decreasing the amount of aggregated forms of both Aß and tau in the brain can serve as potential disease-modifying treatments for AD.


Subject(s)
Alzheimer Disease/drug therapy , Drug Discovery/methods , Thinking , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Humans , Immunotherapy/methods , tau Proteins/antagonists & inhibitors , tau Proteins/metabolism
15.
J Neuropathol Exp Neurol ; 58(9): 1020-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499444

ABSTRACT

The mechanism by which cells die in Alzheimer disease (AD) is unknown. Several investigators speculate that much of the cell loss may be due to apoptosis, a highly regulated form of programmed cell death. Caspase-3 is a critical effector of neuronal apoptosis and may be inappropriately activated in AD. To address this possibility, we examined cortical and hippocampal brain sections from AD patients, as well as 2 animal models of AD, for in situ evidence of caspase-3 activation. We report here that senile plaques and neurofibrillary tangles in the AD brain are not associated with caspase-3 activation. Furthermore, amyloid beta (A beta) deposition in the APPsw transgenic mouse model of AD does not result in caspase-3 activation despite the ability of A beta to induce caspase-3 activation and neuronal apoptosis in vitro. AD brain sections do, however, exhibit caspase-3 activation in hippocampal neurons undergoing granulovacuolar degeneration. Our data suggests that caspase-3 does not have a significant role in the widespread neuronal cell death that occurs in AD, but may contribute to the specific loss of hippocampal neurons involved in learning and memory.


Subject(s)
Alzheimer Disease/enzymology , Caspases/metabolism , Neurons/enzymology , Aged , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/enzymology , Brain/pathology , Caspase 3 , Cells, Cultured , Enzyme Activation , Humans , Immunohistochemistry , Mice , Mice, Transgenic/genetics , Middle Aged , Mutation , Neurofibrillary Tangles/pathology , Peptide Fragments/pharmacology , Plaque, Amyloid/pathology
16.
Stroke ; 34(1): 207-13, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12511776

ABSTRACT

BACKGROUND AND PURPOSE: Different strategies for neuroprotection of neonatal stroke may be required because the developing brain responds differently to hypoxia-ischemia than the mature brain. This study was designed to determine the role of caspase-dependent injury in the pathophysiology of pure focal cerebral ischemia in the immature brain. METHODS: Postnatal day 7 rats were subjected to permanent or transient middle cerebral artery (MCA) occlusion. Diffusion-weighted MRI was used during occlusion to noninvasively map the evolving ischemic core. The time course of caspase-3 activation in ischemic brain tissue was determined with the use of an Asp-Glu-Val-Asp-aminomethylcoumarin cleavage assay. The anatomy of caspase-3 activation in the ischemic core and penumbra was mapped immunohistochemically with an anti-activated caspase-3 antibody in coronal sections that matched the imaging planes on diffusion-weighted MRI. RESULTS: A marked increase in caspase-3 activity occurred within 24 hours of reperfusion after transient MCA occlusion. In contrast, caspase-3 activity remained significantly lower within 24 hours of permanent MCA occlusion. Cells with activated caspase-3 were prominent in the penumbra beginning at 3 hours after reperfusion, while a more delayed but marked caspase-3 activation was observed in the ischemic core by 24 hours after reperfusion. CONCLUSIONS: In the neonate, caspase-3 activation is likely to contribute substantially to cell death not only in the penumbra but also in the core after ischemia with reperfusion. Furthermore, persistent perfusion deficits result in less caspase-3 activation and appear to favor caspase-independent injury.


Subject(s)
Brain Ischemia/enzymology , Caspases/metabolism , Cerebral Cortex/enzymology , Reperfusion Injury/enzymology , Stroke/complications , Animals , Animals, Newborn , Brain/growth & development , Brain Ischemia/etiology , Brain Ischemia/pathology , Caspase 3 , Cerebrovascular Circulation , Diffusion Magnetic Resonance Imaging , Enzyme Activation , Infarction, Middle Cerebral Artery/complications , Kinetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology
17.
J Cereb Blood Flow Metab ; 19(3): 331-40, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10078885

ABSTRACT

Neuroprotection against cerebral ischemia can be realized if the brain is preconditioned by previous exposure to a brief period of sublethal ischemia. The present study was undertaken to test the hypothesis that nitric oxide (NO) produced from the neuronal isoform of NO synthase (NOS) serves as a necessary signal for establishing an ischemia-tolerant state in brain. A newborn rat model of hypoxic preconditioning was used, wherein exposure to sublethal hypoxia (8% oxygen) for 3 hours renders postnatal day (PND) 6 animals completely resistant to a cerebral hypoxic-ischemic insult imposed 24 hours later. Postnatal day 6 animals were treated 0.5 hour before preconditioning hypoxia with the nonselective NOS inhibitor L-nitroarginine (2 mg/kg intraperitoneally). This treatment, which resulted in a 67 to 81% inhibition of calcium-dependent constitutive NOS activity 0.5 to 3.5 hours after its administration, completely blocked preconditioning-induced protection. However, administration of the neuronal NOS inhibitor 7-nitroindazole (40 mg/kg intraperitoneally) before preconditioning hypoxia, which decreased constitutive brain NOS activity by 58 to 81%, was without effect on preconditioning-induced cerebroprotection, as was pretreatment with the inducible NOS inhibitor aminoguanidine (400 mg/kg intraperitoneally). The protective effects of preconditioning were also not blocked by treating animals with competitive [3-(2-carboxypiperazin-4-yl)propyl-1-phosphonate; 5 mg/kg intraperitoneally] or noncompetitive (MK-801; 1 mg/kg intraperitoneally) N-methyl-D-aspartate receptor antagonists prior to preconditioning hypoxia. These findings indicate that NO production and activity are critical to the induction of ischemic tolerance in this model. However, the results argue against the involvement of the neuronal NOS isoform, activated secondary to a hypoxia-induced stimulation of N-methyl-D-aspartate receptors, and against the involvement of the inducible NOS isoform, but rather suggest that NO produced by the endothelial NOS isoform is required to mediate this profound protective effect.


Subject(s)
Brain Ischemia/prevention & control , Hypoxia/physiopathology , Nitric Oxide/physiology , Animals , Animals, Newborn , Calcium/pharmacology , Dizocilpine Maleate/pharmacology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Indazoles/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitroarginine/pharmacology , Oxygen/administration & dosage , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology
18.
Arch Neurol ; 45(9): 945-8, 1988 Sep.
Article in English | MEDLINE | ID: mdl-2843154

ABSTRACT

We prospectively studied 40 hospitalized patients who had well-established diagnoses of acquired immunodeficiency syndrome. Patients with confounding risk factors for neuropathy were excluded; none of the study patients had known vitamin deficiency, alcoholism, or any metabolic, drug, or toxic factor. Clinical and electrophysiologic evidence of a distal symmetric polyneuropathy was found in 35% (13/37) of the patients. Symptoms and signs of neuropathy were usually mild, and painful dysesthesias were uncommon. Amplitude reduction of sural nerve action potentials distinguished all patients with from those without clinical neuropathy. Results of other electrophysiologic studies of sural, peroneal, and median nerves were typically normal. These results provide evidence of distal axonal degeneration. Neuropathy occurred only in patients with systemic illness longer than five months' duration. When compared with patients without neuropathy, these patients had more severe weight loss and a higher incidence of clinical dementia. Follow-up evaluation showed no evidence of clinical progression over a six-month period. The pathogenesis of this common distal axonal polyneuropathy is unknown and warrants further investigation.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Peripheral Nervous System Diseases/complications , Acquired Immunodeficiency Syndrome/mortality , Action Potentials , Adult , Dementia/complications , Electrophysiology , Follow-Up Studies , Humans , Male , Neural Conduction , Peripheral Nervous System Diseases/physiopathology , Prospective Studies , Sural Nerve
19.
Neurology ; 43(12): 2668-73, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8255474

ABSTRACT

Atrophy and dysfunction of certain neurons, including cholinergic neurons in the basal forebrain, are key features of the neuropathology of Alzheimer's disease (AD). Since all individuals with Down syndrome (DS) develop AD neuropathology by the 4th decade, we reasoned that a genetic model of DS, the trisomy 16 (Ts 16) mouse, may provide an animal model to study the neurodegeneration in AD. Ts 16 mice fail to survive birth; to evaluate neurons for long periods in vivo required transplantation of fetal tissue. We previously demonstrated that Ts 16 basal forebrain cholinergic neurons (BFCNs) undergo age-related atrophy similar to DS and AD, and now show that a specific neurotrophic factor, nerve growth factor (NGF), acts to reverse Ts 16-induced atrophy of BFCNs and stimulates hypertrophy of these cells. As NGF levels were not decreased in the host, abnormalities intrinsic to Ts 16 BFCNs presumably caused the atrophy. Our results suggest that NGF may be useful in reversing cholinergic neurodegeneration in DS and AD.


Subject(s)
Aging/physiology , Down Syndrome/pathology , Nerve Degeneration/drug effects , Nerve Growth Factors/pharmacology , Neurons/drug effects , Animals , Atrophy , Brain/pathology , Cell Transplantation , Disease Models, Animal , Fetal Tissue Transplantation , Hippocampus/physiopathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neurons/pathology , Prosencephalon/embryology
20.
Am J Med ; 87(2): 173-7, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2757058

ABSTRACT

PURPOSE: We attempt to define the significance and most common causes of new-onset seizures in patients with human immunodeficiency virus (HIV) infection. In addition, we review the seizure type, neurologic examination, and other clinical features to better address diagnostic and management issues in these patients. PATIENTS AND METHODS: We reviewed 100 cases of new-onset seizures in HIV-infected patients who underwent complete evaluations at the University of California, San Francisco, hospitals. RESULTS: Seizures were the presenting symptom of HIV-related illness in 18 patients, six of whom developed no other HIV-related illness until at least four months after the first seizure. Common causes in the 100 patients included mass lesions, HIV encephalopathy, and meningitis. No cause for the seizures was found in 23 patients despite a complete evaluation. An underlying cause was found in all patients with focal neurologic deficits but in only two of 24 who had normal results on an interictal neurologic examination. Focal ictal features were not predictive of cause. A cause was found in all 12 patients with status epilepticus or medically refractory seizures. A total of 12 of the 87 (14%) patients who received phenytoin developed a hypersensitivity reaction. Despite the brevity of follow-up in some patients, many patients, including those with no definable cause, had multiple seizures prior to the administration of anticonvulsants. CONCLUSION: The direct effects of HIV on the brain may be the single most common cause of seizures in this population. We favor treatment of a single seizure in patients with HIV infection.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Seizures/etiology , Adult , Brain Diseases/cerebrospinal fluid , Brain Diseases/diagnosis , Brain Diseases/etiology , Electroencephalography , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Phenytoin/adverse effects , Phenytoin/therapeutic use , Seizures/drug therapy , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL