Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Neuropathol Appl Neurobiol ; 39(6): 654-66, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23231074

ABSTRACT

AIMS: Traumatic brain injury is a significant cause of morbidity and mortality worldwide. An epidemiological association between head injury and long-term cognitive decline has been described for many years and recent clinical studies have highlighted functional impairment within 12 months of a mild head injury. In addition chronic traumatic encephalopathy is a recently described condition in cases of repetitive head injury. There are shared mechanisms between traumatic brain injury and Alzheimer's disease, and it has been hypothesized that neuroinflammation, in the form of microglial activation, may be a mechanism underlying chronic neurodegenerative processes after traumatic brain injury. METHODS: This study assessed the microglial reaction after head injury in a range of ages and survival periods, from <24-h survival through to 47-year survival. Immunohistochemistry for reactive microglia (CD68 and CR3/43) was performed on human autopsy brain tissue and assessed 'blind' by quantitative image analysis. Head injury cases were compared with age matched controls, and within the traumatic brain injury group cases with diffuse traumatic axonal injury were compared with cases without diffuse traumatic axonal injury. RESULTS: A major finding was a neuroinflammatory response that develops within the first week and persists for several months after traumatic brain injury, but has returned to control levels after several years. In cases with diffuse traumatic axonal injury the microglial reaction is particularly pronounced in the white matter. CONCLUSIONS: These results demonstrate that prolonged microglial activation is a feature of traumatic brain injury, but that the neuroinflammatory response returns to control levels after several years.


Subject(s)
Brain Injuries/immunology , Brain/immunology , Microglia/immunology , Adolescent , Adult , Age Factors , Aged , Brain Injuries/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Microglia/pathology , Middle Aged , Young Adult
2.
Neuroscience ; 151(1): 104-10, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18068305

ABSTRACT

Recent data suggest that the endocannabinoid system (ECS) may be involved in the glial response in different types of brain injury. Both acute and chronic insults seem to trigger a shift in the pattern of expression of some elements of this system from neuronal to glial. Specifically, data obtained in human brain tissue sections from Alzheimer's disease patients showed that the expression of cannabinoid receptors of the CB(2) type is induced in activated microglial cells while fatty acid amide hydrolase (FAAH) expression is increased in reactive astrocytes. The present study was designed to determine the time-course of the shift from neuronal to glial induction in the expression of these proteins in Down's syndrome, sometimes referred to as a human model of Alzheimer-like beta-amyloid (Abeta) deposition. Here we present immunohistochemical evidence that both CB(2) receptors and FAAH enzyme are induced in Abeta plaque-associated microglia and astroglia, respectively, in Down's syndrome. These results suggest that the induction of these elements of the ECS contributes to, or is a result of, amyloid deposition and subsequent plaque formation. In addition, they confirm a striking differential pattern of distribution of FAAH and CB(2) receptors.


Subject(s)
Amidohydrolases/metabolism , Amyloid beta-Peptides/physiology , Down Syndrome/metabolism , Neuroglia/metabolism , Receptor, Cannabinoid, CB2/metabolism , Adult , Brain/pathology , Brain Chemistry/physiology , Child , Down Syndrome/pathology , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Infant, Newborn , Male , Middle Aged , Plaque, Amyloid/pathology
3.
Neuropathol Appl Neurobiol ; 31(3): 270-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15885064

ABSTRACT

Argyrophilic grain disease (AGD) constitutes a neurodegenerative disorder that occurs in the brains of the elderly and affects 5% of all patients with dementia. Tau protein-containing lesions known as argyrophilic grains and located predominantly in limbic regions of the brain characterize this disease. Dementia is encountered in only a subset of cases that display the morphological pattern of AGD. The aim of this study is to determine the role of concurrent Alzheimer's disease (AD)-related pathology for the development of dementia in AGD patients. A total of 204 post-mortem brains from 30 demented and 49 nondemented AGD patients, 39 AD patients, and from 86 nondemented controls without AGD were staged for AD-related neurofibrillary tangles (NFTs) as well as amyloid beta-protein (Abeta) deposition. To identify differences in AD-related pathology between demented and nondemented AGD cases, and to differentiate the pattern of AD-related changes in demented and nondemented AGD cases from that seen in AD and nondemented controls, we statistically compared the stages of Abeta and NFT distribution among these groups. Using a logistic regression model, we showed that AGD has a significant effect on the development of dementia beyond that attributable to AD-related pathology (P < 0.005). Demented AGD cases showed lower stages of AD-related pathology than did pure AD cases but higher stages than nondemented AGD patients. AGD associated dementia was seen in the presence of NFT (Braak)-stages II-IV and Abeta-phases 2-3, whereas those stages were not associated with dementia in the absence of AGD. In conclusion, AGD is a clinically relevant neurodegenerative entity that significantly contributes to the development of dementia by lowering the threshold for cognitive deficits in the presence of moderate amounts of AD-related pathology.


Subject(s)
Alzheimer Disease/complications , Brain/pathology , Inclusion Bodies/pathology , Neurodegenerative Diseases/complications , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Female , Humans , Logistic Models , Male , Middle Aged , Neurodegenerative Diseases/pathology , Neurofibrillary Tangles/pathology
4.
J Neuropsychiatry Clin Neurosci ; 17(4): 541-3, 2005.
Article in English | MEDLINE | ID: mdl-16387995

ABSTRACT

Previous studies have reported associations between apolipoprotein E (APOE) genotype, cognitive function, and psychopathology in psychiatric populations. The authors investigated the associations between APOE allele status, memory function, and posttraumatic stress disorder (PTSD) symptom severity in PTSD subjects. Presence of the APOE 2 allele was associated with significantly worse reexperiencing symptoms and impaired memory function in this population.


Subject(s)
Alleles , Apolipoproteins E/genetics , Combat Disorders/genetics , Memory Disorders/etiology , Apolipoproteins E/classification , Combat Disorders/physiopathology , Humans , Male , Middle Aged , Psychiatric Status Rating Scales/statistics & numerical data
5.
Forensic Sci Int ; 146(2-3): 97-104, 2004 Dec 16.
Article in English | MEDLINE | ID: mdl-15542269

ABSTRACT

Epidemiological and pathological studies suggest that head injury is a significant risk factor for subsequent neurodegeneration and cognitive decline in later life. The precise mechanisms for the development of post-traumatic neurodegenerative change are unclear but we hypothesize that persistence of inflammatory processes in the brain may play a key role and that some individuals are more susceptible to such changes based on their genetic make-up. In support of this hypothesis we present evidence of persistent elevated microglial activity in long-term survivors of head injury and the suggestion of an association between the extent of this activity and interleukin-1 genotype.


Subject(s)
Brain Injuries/pathology , Brain/pathology , Inflammation/pathology , Microglia/pathology , Adolescent , Adult , Aged , Alleles , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apolipoproteins E/genetics , Biomarkers/metabolism , Brain/metabolism , Brain Injuries/metabolism , Case-Control Studies , Child , Child, Preschool , Female , Forensic Pathology , Genotype , Humans , Hyperplasia/metabolism , Hypertrophy/metabolism , Infant , Inflammation/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Macrophage-1 Antigen/metabolism , Male , Microglia/metabolism , Middle Aged , Phagocytosis , Survival Analysis
6.
Neurochem Int ; 39(5-6): 341-8, 2001.
Article in English | MEDLINE | ID: mdl-11578769

ABSTRACT

Activated (phosphorylated) mitogen-activated protein kinase p38 (MAPK-p38) and interleukin-1 (IL-1) have both been implicated in the hyperphosphorylation of tau, a major component of the neurofibrillary tangles in Alzheimer's disease. This, together with findings showing that IL-1 activates MAPK-p38 in vitro and is markedly overexpressed in Alzheimer brain, suggest a role for IL-1-induced MAPK-p38 activation in the genesis of neurofibrillary pathology in Alzheimer's disease. We found frequent colocalization of hyperphosphorylated tau protein (AT8 antibody) and activated MAPK-p38 in neurons and in dystrophic neurites in Alzheimer brain, and frequent association of these structures with activated microglia overexpressing IL-1. Tissue levels of IL-1 mRNA as well as of both phosphorylated and non-phosphorylated isoforms of tau were elevated in these brains. Significant correlations were found between the numbers of AT8- and MAPK-p38-immunoreactive neurons, and between the numbers of activated microglia overexpressing IL-1 and the numbers of both AT8- and MAPK-p38-immunoreactive neurons. Furthermore, rats bearing IL-1-impregnated pellets showed a six- to seven-fold increase in the levels of MAPK-p38 mRNA, compared with rats with vehicle-only pellets (P<0.0001). These results suggest that microglial activation and IL-1 overexpression are part of a feedback cascade in which MAPK-p38 overexpression and activation leads to tau hyperphosphorylation and neurofibrillary pathology in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Interleukin-1/physiology , Mitogen-Activated Protein Kinases/metabolism , Aged , Aged, 80 and over , Animals , Blotting, Western , Brain/enzymology , Female , Humans , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-1/pharmacology , Male , Middle Aged , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases , tau Proteins/metabolism
7.
Neurochem Int ; 39(5-6): 409-13, 2001.
Article in English | MEDLINE | ID: mdl-11578776

ABSTRACT

Postmortem demonstration of increased expression of biologically active S100B in Alzheimer's disease (AD) and its relation to progression of neuropathological changes across the cortical regions suggests involvement of this astrocytic cytokine in the pathophysiology of AD. The hypothesis that the overexpression of S100B in Alzheimer brain is related to the progression of clinical symptoms was addressed in living persons by measuring S100B concentrations in cerebrospinal fluid (CSF) from AD patients with a broad range of clinical dementia severity and from healthy older persons. The effect of normal aging on CSF S100B concentrations also was estimated. CSF S100B did not differ between all 68 AD subjects (0.98+/-0.09 ng/ml (mean+/-S.E.M.)) and 25 healthy older subjects (0.81+/-0.13 ng/ml). When AD subjects were divided into mild/moderate stage and advanced stage clinical dementia severity by the established Clinical Dementia Rating Scale (CDR) criteria, S100B was significantly higher in the 46 mild/moderate stage AD subjects (1.17+/-0.11 ng/ml) than in either the 22 advanced stage AD subjects (0.60+/-0.12 ng/ml) or the healthy older subjects. Consistent with higher CSF S100B in mild to moderate AD, there was a significant correlation among all AD subjects between CSF S100B and cognitive status as measured by the Mini Mental State Exam (MMSE) score. CSF S100B did not differ between healthy older subjects and healthy young subjects. These results suggest increased CNS expression of S100B in the earlier stages of AD, and are consistent with a role for S100B in the initiation and/or facilitation of neuritic plaque formation in AD brain.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Calcium-Binding Proteins/cerebrospinal fluid , Nerve Growth Factors/cerebrospinal fluid , S100 Proteins , Adult , Aged , Alzheimer Disease/psychology , Cognition , Female , Humans , Male , Mental Status Schedule , Psychiatric Status Rating Scales , Reference Values , S100 Calcium Binding Protein beta Subunit , Time Factors
8.
J Neurosci Res ; 66(2): 163-70, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11592111

ABSTRACT

Neurotoxic microglial-neuronal interactions have been implicated in the pathogenesis of various neurodegenerative diseases such as Alzheimer's disease, and vitamin E has been shown to have direct neuroprotective effects. To determine whether vitamin E also has indirect neuroprotective effects through suppression of microglial activation, we used a microglial-neuronal coculture. Lipopolysaccharide (LPS) treatment of a microglial cell line (N9) induced a time-dependent activation of both p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor-kappaB (NFkappaB), with consequent increases in interleukin-1alpha (IL-1alpha), tumor necrosis factor-alpha (TNF-alpha), and nitric oxide (NO) production. Differentiated neuronal cells (PC12 cells treated with nerve growth factor) exhibited marked loss of processes and decreased survival when cocultured with LPS-activated microglia. Preincubation of microglia with vitamin E diminished this neurotoxic effect, independently of direct effects of the antioxidant on the neuronal cells. Microglial NO production and the induction of IL-1alpha and TNFalpha expression also were attenuated by vitamin E. Such antiinflammatory effects of vitamin E were correlated with suppression of p38 MAPK and NFkappaB activation and were mimicked by an inhibition of either p38 MAPK (by SB203580) or NFkappaB (by decoy oligonucleotides). These results suggest that, in addition to the beneficial effects of providing direct antioxidant protection to neurons reported by others, vitamin E may provide neuroprotection in vivo through suppression of signaling events necessary for microglial activation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cytokines/metabolism , Microglia/drug effects , Neuroprotective Agents/pharmacology , Vitamin E/pharmacology , Animals , Antioxidants/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Coculture Techniques , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Interleukin-1/biosynthesis , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Mice , Microglia/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/biosynthesis , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , PC12 Cells/pathology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Pyridines/pharmacology , Rats , Thionucleotides/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases
9.
Psychiatry Res ; 106(3): 171-80, 2001 May 30.
Article in English | MEDLINE | ID: mdl-11382539

ABSTRACT

It has been hypothesized that schizophrenia arises from cell membrane abnormalities due to changes in phospholipid (PL) composition and metabolism. We have used high resolution, in vitro 31P nuclear magnetic resonance (NMR) to characterize the PLs in left frontal cortex (gray matter) of postmortem brain from four schizophrenics and five controls. High resolution 31P NMR spectra were obtained in an organic-solvent system to resolve PL classes (headgroups) and in a sodium-cholate, aqueous dispersion system to resolve phosphatidylcholine (PC) molecular species. Multivariate analysis which included the major PC molecular species and phosphatidylinositol (PI) showed a significant difference between schizophrenics and controls. Analysis of specific interactions showed that the PI was significantly higher in the schizophrenic group than in the control group. There were no differences between the two groups for other individual PL classes, or for individual PL subclasses determined by the linkage type at the sn-1 position on glycerol. There was a trend for total PL content to be higher in schizophrenics than in controls. There was no evidence for elevated lysophosphatidylcholine or lysophosphatidylethanolamine in schizophrenia. The intensity of the PC peak representing molecular species with one saturated and one unsaturated (one or two double bonds) acyl chain was higher for the schizophrenic group than for the control group. Although these results are not in complete agreement with previous studies, they support the idea that PL abnormalities occur in the brain in schizophrenia and that fatty acid metabolism may be abnormal.


Subject(s)
Brain/metabolism , Magnetic Resonance Spectroscopy , Phospholipids/metabolism , Schizophrenia/metabolism , Adult , Aged , Aged, 80 and over , Female , Frontal Lobe/metabolism , Humans , Male , Middle Aged , Phosphorus
10.
Neurobiol Aging ; 22(6): 903-8, 2001.
Article in English | MEDLINE | ID: mdl-11754997

ABSTRACT

Interleukin-1 (IL-1)-1) is a pluripotent immunomodulatory cytokine that has an initiating role in cellular and humoral immunity in the periphery. Il-1 is overexpressed in Alzheimer brain, and this overexpression is directly related to plaque formation and progression, nonsensical growth of dystrophic neurites, and neuronal overexpression of acetylcholinesterase. IL-1 has a number of actions relevant to Alzheimer's disease, including excessive expression of neuronal Abeta precursor protein and other plaque-associated proteins, and induction of astrocyte activation and astrocytic overexpression of S100B. These latter events may be related to the overgrowth of dystrophic neurites in neuritic plaques, a necessary event for conversion of diffuse Abeta deposits into the neuritic amyloid plaques diagnostic of Alzheimer's disease. Four new genetic studies underscore the relevance of IL-1 to Alzheimer pathogenesis, showing that homozygosity of a specific polymorphism in the IL-1A gene at least triples Alzheimer risk, especially for an earlier age of onset and in combination with homozygosity for another polymorphism in the IL-1B gene.


Subject(s)
Alzheimer Disease/pathology , Inflammation/pathology , Interleukin-1/physiology , Neurons/pathology , Alzheimer Disease/genetics , Animals , Brain Chemistry/genetics , Brain Chemistry/physiology , Humans , Inflammation/genetics , Interleukin-1/genetics , Polymorphism, Genetic/genetics
11.
Neurobiol Aging ; 22(6): 895-902, 2001.
Article in English | MEDLINE | ID: mdl-11754996

ABSTRACT

Transgenic APPV717F mice, homozygous for a human minigene encoding the V717F familial Alzheimer's disease mutation, develop Abeta plaques similar to those seen in Alzheimer patients and show evidence of neuronal cell drop out in CA2-3 regions of the hippocampus at 8 months of age and older. Interleukin-1 (IL-1)beta (IL-1beta) converting enzyme (ICE) is a cysteine protease (caspase-1) that processes inactive (33 kDa) pro-IL-1beta to the active (17 kDa) inflammatory cytokine. We used immunohistochemistry, RT-PCR, and DNA cleavage (TUNEL) analysis to show progressive, age-associated increases in ICE mRNA levels, in the numbers of ICE-immunoreactive glia, and in the numbers of neurons showing evidence of DNA damage in APPV717F mice that commenced months prior to the appearance of Abeta plaques. Moreover, there were significant correlations between these parameters over an age range of 1-17 months. These findings are consistent with the idea that increases in ICE activity and expression contribute to neuronal injury in Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Caspase 1/biosynthesis , Caspase 1/genetics , DNA Damage/physiology , Gene Expression Regulation, Enzymologic/genetics , Neurons/metabolism , Animals , Female , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Transgenic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Silver Staining
12.
Exp Gerontol ; 35(4): 481-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10959036

ABSTRACT

Interleukin-1 (IL-1) has been implicated as a key molecule in Alzheimer pathogenesis based on findings of an IL-1 overexpression in Alzheimer brain that is directly related to plaque progression and tangle formation, and on findings that IL-1 induces excessive synthesis, translation, and processing of neuronal beta-amyloid precursor protein (betaAPP) as well as synthesis of most known plaque-associated proteins. In addition, IL-1 activates astrocytes, with the important consequence of overexpression of the neuritogenic cytokine S100beta and overgrowth of dystrophic neurites in neuritic plaques. As further evidence of the importance of IL-1 in Alzheimer pathogenesis, two new genetic studies of inheritance of specific polymorphisms in IL-1 genes in Alzheimer and control patients show that homozygosity for a specific IL-1A gene polymorphism at least triples risk for development of Alzheimer's disease. This increase is associated with earlier age of onset. Homozygosity for this polymorphism plus another in the IL-1B gene further increases risk.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Genetic Predisposition to Disease , Interleukin-1/genetics , Interleukin-1/metabolism , Polymorphism, Genetic , Humans
13.
Neurology ; 55(4): 480-3, 2000 Aug 22.
Article in English | MEDLINE | ID: mdl-10953177

ABSTRACT

BACKGROUND: Retrospective epidemiologic studies suggest that individuals exposed to anti-inflammatory agents such as nonsteroidal anti-inflammatory drugs have a lower probability of developing AD as well as an older age at onset for the illness. Neuroinflammation may play an important role in the pathogenesis of AD. Interleukin 1 (IL-1), a potent proinflammatory cytokine, is colocalized immunohistochemically to neuritic plaques, a requisite neuropathologic feature for AD. A polymorphism in the 5'-flanking regulatory region at -889 of the IL-1 alpha gene (a C-to-T transition designated as IL-1A[-889] allele 2) may cause an overexpression of IL-1 alpha, a finding shown to be associated with inflammatory diseases. The IL-1A(-889) allele 2 polymorphism may be associated with AD pathogenesis. METHODS: A total of 259 patients with AD and 192 nondemented control subjects were included from two different centers (Indianapolis, IN, and Munich, Germany). Genotyping for APOE alleles and IL-1A(-889) allele 2 was performed by PCR-based amplification followed by restrictive endonuclease digestion. Statistical analyses were conducted by center-, gender group-, and age group-stratified Mantel-Haenszel odds ratios, CI, and p values. RESULTS: The allele frequency of IL-1A(-889) allele 2 was 46% in clinically diagnosed patients with probable AD versus 34% in control subjects from the combined centers. CONCLUSION: The authors found an increased risk for AD with an estimated Mantel-Haenszel odds ratio of 1.68 (95% CI 1.1 to 2.6; p = 0.022) for heterozygous carriers and 7.2 (95% CI 2.0 to 24.5; p = 0.003) for individuals homozygous for IL-1A(-889) allele 2. They found no evidence for an interaction between the IL-1A and the apoE epsilon 4 polymorphisms (carriers and homozygotes), age, or gender with regard to conferred risk. The data strongly support an association between the IL-1A(-889) allele 2, especially in homozygotes, and later-onset AD.


Subject(s)
Alzheimer Disease/genetics , Interleukin-1/genetics , Polymorphism, Genetic/genetics , Aged , Aged, 80 and over , Alleles , Alzheimer Disease/immunology , Apolipoprotein E4 , Apolipoproteins E/genetics , Case-Control Studies , Cohort Studies , DNA Mutational Analysis , Female , Gene Frequency , Genetic Carrier Screening , Genotype , Heterozygote , Homozygote , Humans , Logistic Models , Male , Middle Aged , Odds Ratio , Risk Assessment
14.
Exp Neurol ; 163(2): 388-91, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10833312

ABSTRACT

Slow-release, IL-1-impregnated pellets implanted in rat cerebral cortex to simulate chronic overexpression of IL-1 significantly increased relative tissue levels of tau mRNA and of tau immunoreactivity in neuronal cell bodies and in swollen dystrophic neurites that also overexpressed phosphorylated and nonphosphorylated neurofilament epitopes. In addition, rats with IL-1-impregnated pellets, but not control rats or those with sham pellets, showed focal immunoreactivity for hyperphosphorylated tau epitopes present in paired helical filaments. Our results are consistent with an important driving role for IL-1 in the pathogenesis of Alzheimer-type neuronal and neuritic changes.


Subject(s)
Interleukin-1/pharmacology , Neurofilament Proteins/drug effects , tau Proteins/drug effects , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Delayed-Action Preparations , Drug Implants , Hippocampus/drug effects , Hippocampus/metabolism , Interleukin-1/metabolism , Male , Neurofilament Proteins/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , tau Proteins/metabolism
15.
Neurobiol Aging ; 21(3): 383-421, 2000.
Article in English | MEDLINE | ID: mdl-10858586

ABSTRACT

Inflammation clearly occurs in pathologically vulnerable regions of the Alzheimer's disease (AD) brain, and it does so with the full complexity of local peripheral inflammatory responses. In the periphery, degenerating tissue and the deposition of highly insoluble abnormal materials are classical stimulants of inflammation. Likewise, in the AD brain damaged neurons and neurites and highly insoluble amyloid beta peptide deposits and neurofibrillary tangles provide obvious stimuli for inflammation. Because these stimuli are discrete, microlocalized, and present from early preclinical to terminal stages of AD, local upregulation of complement, cytokines, acute phase reactants, and other inflammatory mediators is also discrete, microlocalized, and chronic. Cumulated over many years, direct and bystander damage from AD inflammatory mechanisms is likely to significantly exacerbate the very pathogenic processes that gave rise to it. Thus, animal models and clinical studies, although still in their infancy, strongly suggest that AD inflammation significantly contributes to AD pathogenesis. By better understanding AD inflammatory and immunoregulatory processes, it should be possible to develop anti-inflammatory approaches that may not cure AD but will likely help slow the progression or delay the onset of this devastating disorder.


Subject(s)
Alzheimer Disease/pathology , Inflammation/pathology , Brain/pathology , Humans
16.
J Neuropathol Exp Neurol ; 59(6): 471-6, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10850859

ABSTRACT

Established genetic causes of familial Alzheimer disease (AD) involve genes for beta-amyloid precursor protein (betaAPP), presenilin-1, and presenilin-2. For the more common sporadic forms of AD, increased risk has been associated with a number of genes; the most important of which is the epsilon4 allele of apolipoprotein E. Two recent studies, one clinical and one using postmortem material, now show increased risk for AD associated with certain polymorphisms in the genes encoding the alpha and beta isoforms of interleukin-1 (IL-1). IL-1 levels are elevated in Alzheimer brain, and overexpression of IL-1 is associated with beta-amyloid plaque progression. IL-1 interacts with the gene products of several other known or suspected genetic risk factors for AD, including betaAPP, apolipoprotein E, alpha1-antichymotrypsin, and alpha2-macroglobulin. IL-1 overexpression is also associated with environmental risk factors for AD, including normal aging and head trauma. These observations suggest an important pathogenic role for IL-1, and for IL-1-driven cascades, in the pathogenesis of AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/immunology , Genetic Predisposition to Disease , Interleukin-1/genetics , Alzheimer Disease/etiology , Alzheimer Disease/physiopathology , Humans , Immunogenetics , Interleukin-1/physiology , Polymorphism, Genetic/physiology , Risk Factors
17.
Ann Neurol ; 47(3): 361-5, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10716256

ABSTRACT

Overexpression of the pluripotent cytokine interleukin-1 (IL-1) by microglial cells correlates with formation of neuritic beta-amyloid plaques in Alzheimer's disease (AD). We evaluated polymorphisms in the genes coding for the IL-1alpha, IL-1beta, and IL-1 receptor antagonist cytokines, and tested their association with the occurrence and age at onset of sporadic AD. We found a strong association between the IL-1A T/T genotype and AD onset before 65 years of age (odds ratio, 4.86), with carriers of this genotype showing an onset of disease 9 years earlier than IL-1A C/C carriers. A weaker association with the age at onset was also shown for the IL-1B and IL-1RN genes. These data suggest either a direct effect of the IL-1 gene family, mainly IL-1A, on the clinical onset of AD, or a linkage dysequilibrium with an unknown locus relevant to AD on chromosome 2.


Subject(s)
Alzheimer Disease/genetics , Interleukin-1/genetics , Polymorphism, Genetic/genetics , Adult , Age of Onset , Aged , Aged, 80 and over , Alzheimer Disease/etiology , Female , Genotype , Humans , Male , Middle Aged , Risk Factors
18.
Ann Neurol ; 47(3): 365-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10716257

ABSTRACT

Interleukin-1 (IL-1) is markedly overexpressed in Alzheimer's disease. We found the IL-1A 2,2 genotype in 12.9% of 232 neuropathologically confirmed Alzheimer's disease patients and 6.6% of 167 controls from four centers in the United Kingdom and United States (odds ratio, 3.0; controlled for age and for ApoE [apolipoprotein E] genotype). Homozygosity for both allele 2 of IL-1A and allele 2 of IL-1B conferred even greater risk (odds ratio, 10.8). IL-1 genotypes may confer risk for Alzheimer's disease through IL-1 overexpression and IL-1-driven neurodegenerative cascades.


Subject(s)
Alzheimer Disease/genetics , Interleukin-1/genetics , Polymorphism, Genetic/genetics , Aged , Aged, 80 and over , Alleles , Genotype , Humans
19.
J Neurochem ; 74(1): 143-50, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10617115

ABSTRACT

Levels of the neurotrophic cytokine S100beta and the proinflammatory cytokine interleukin-6 (IL-6) are both elevated in Alzheimer's brain, and both have been implicated in beta-amyloid plaque formation and progression. We used RT-PCR and electrophoretic mobility shift assay to assess S100beta induction of IL-6 expression and the role of kappaB-dependent transcription in this induction in neuron-enriched cultures and in neuron-glia mixed cultures from fetal rat cortex. S100beta (10 or 100 ng/ml x 24 h) increased IL-6 mRNA levels two- and fivefold, respectively (p<0.05 in each case), and S100beta (100-1,000 ng/ml) induced increases in medium levels of biologically active IL-6 (30-80%). Combined in situ hybridization and immunohistochemistry preparations localized IL-6 mRNA to neurons in these cultures. S100beta induction of IL-6 expression correlated with an increase in DNA binding activity specific for a KB element and was inhibited (75%) by suppression of kappaB binding with double-stranded "decoy" oligonucleotides. The low levels of S100beta required to induce IL-6 overexpression in neurons, shown here, suggest that overexpression of S100beta induces neuronal expression of IL-6 and of IL-6-induced neurodegenerative cascades in Alzheimer's disease.


Subject(s)
Inflammation Mediators/metabolism , Interleukin-6/metabolism , Neurons/drug effects , Neurons/metabolism , S100 Proteins/pharmacology , Animals , Cells, Cultured , Coculture Techniques , Interleukin-6/genetics , Interleukin-6/physiology , NF-kappa B/metabolism , Neuroglia/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
20.
J Neurochem ; 74(1): 295-301, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10617132

ABSTRACT

Homozygous APPV717F transgenic mice overexpress a human beta-amyloid precursor protein (betaAPP) minigene encoding a familial Alzheimer's disease mutation. These mice develop Alzheimer-type neuritic beta-amyloid plaques surrounded by astrocytes. S100beta is an astrocyte-derived cytokine that promotes neurite growth and promotes excessive expression of betaAPP. S100beta overexpression in Alzheimer's disease correlates with the proliferation of betaAPP-immunoreactive neurites in beta-amyloid plaques. We found age-related increases in tissue levels of both betaAPP and S100beta mRNA in transgenic mice. Neuronal betaAPP overexpression was found in cell somas in young mice, whereas older mice showed betaAPP overexpression in dystrophic neurites in plaques. These age-related changes were accompanied by progressive increases in S100beta expression, as determined by S100beta load (percent immunoreactive area). These increases were evident as early as 1 and 2 months of age, months before the appearance of beta-amyloid deposits in these mice. Such precocious astrocyte activation and S100beta overexpression are similar to our earlier findings in Down's syndrome. Accelerated age-related overexpression of S100beta may interact with age-associated overexpression of mutant betaAPP in transgenic mice to promote development of Alzheimer-like neuropathological changes.


Subject(s)
Neurites/physiology , Aging/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Astrocytes/metabolism , Astrocytes/physiology , Female , Glial Fibrillary Acidic Protein/genetics , Hippocampus/cytology , Hippocampus/metabolism , Humans , Immunohistochemistry , Mice , Mice, Transgenic/genetics , Plaque, Amyloid/metabolism , RNA, Messenger/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...