Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 296: 100801, 2021.
Article in English | MEDLINE | ID: mdl-34019878

ABSTRACT

Phagocytosis plays diverse roles in biology, but our understanding of the purpose, interplay, and cell signaling mechanisms associated with different modes of phagocytosis is limited, without being able to capture and visualize each step in this rapid process from the beginning to end. A new study by Walbaum et al. uses stunning time-lapse 3D imaging of the engulfment of erythrocytes by macrophages via sinking, ruffling, and cup formation, unequivocally confirming a visionary 44-year-old theory derived from still electron microscopy photos that phagocytosis mediated by complement receptor CR3 occurs via a sinking mechanism and antibody-mediated phagocytosis occurs via phagocytic cup formation. The article also challenges the dogma, showing that phagocytic cup formation is not unique to antibody receptor phagocytosis, rather CR3 plays a complex role in different modes of phagocytosis. For example, inhibition of antibody-mediated phagocytosis leads to a compensatory upregulation of CR3-mediated sinking phagocytosis. These findings animate, in vivid colors, processes previously only captured as stills, exposing interactions between different phagocytic mechanisms and altering our basic understanding of this important process.


Subject(s)
Phagocytes/metabolism , Receptors, Complement/metabolism , Receptors, IgG/metabolism , Animals , Complement System Proteins/physiology , Phagocytosis/physiology
2.
Neurobiol Dis ; 127: 432-448, 2019 07.
Article in English | MEDLINE | ID: mdl-30951849

ABSTRACT

Alzheimer's disease (AD) genetics implies a causal role for innate immune genes, TREM2 and CD33, products that oppose each other in the downstream Syk tyrosine kinase pathway, activating microglial phagocytosis of amyloid (Aß). We report effects of low (Curc-lo) and high (Curc-hi) doses of curcumin on neuroinflammation in APPsw transgenic mice. Results showed that Curc-lo decreased CD33 and increased TREM2 expression (predicted to decrease AD risk) and also increased TyroBP, which controls a neuroinflammatory gene network implicated in AD as well as phagocytosis markers CD68 and Arg1. Curc-lo coordinately restored tightly correlated relationships between these genes' expression levels, and decreased expression of genes characteristic of toxic pro-inflammatory M1 microglia (CD11b, iNOS, COX-2, IL1ß). In contrast, very high dose curcumin did not show these effects, failed to clear amyloid plaques, and dysregulated gene expression relationships. Curc-lo stimulated microglial migration to and phagocytosis of amyloid plaques both in vivo and in ex vivo assays of sections of human AD brain and of mouse brain. Curcumin also reduced levels of miR-155, a micro-RNA reported to drive a neurodegenerative microglial phenotype. In conditions without amyloid (human microglial cells in vitro, aged wild-type mice), Curc-lo similarly decreased CD33 and increased TREM2. Like curcumin, anti-Aß antibody (also reported to engage the Syk pathway, increase CD68, and decrease amyloid burden in human and mouse brain) increased TREM2 in APPsw mice and decreased amyloid in human AD sections ex vivo. We conclude that curcumin is an immunomodulatory treatment capable of emulating anti-Aß vaccine in stimulating phagocytic clearance of amyloid by reducing CD33 and increasing TREM2 and TyroBP, while restoring neuroinflammatory networks implicated in neurodegenerative diseases.


Subject(s)
Alzheimer Disease/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain/drug effects , Curcumin/pharmacology , Gene Expression/drug effects , Immunity, Innate/genetics , Microglia/drug effects , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Disease Progression , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Microglia/metabolism , Phagocytosis/drug effects , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Receptors, Immunologic/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism
3.
Curr Drug Targets CNS Neurol Disord ; 4(5): 469-80, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16266282

ABSTRACT

This review will focus primarily on the role of the low density lipoprotein receptor-related protein (LRP-1) in neuronal synapse formation and function in Alzheimer's Disease (AD). We review the role that its ligands may have in cognition or AD: apolipoprotein E (ApoE), alpha2-macroglobulin, Transforming Growth Factor-Beta (TGFbeta, Tissue Plasminogen Activator (tPA), insulin growth factor binding protein-3 (IGFBP-3), which all bind LRP-1 and apolipoprotein J (ApoJ), which is a ligand for LRP-2. After reviewing its role as a signaling receptor, we discuss the connection between LRP and the NMDA glutamate receptor via the post synaptic density 95 (PSD-95) neuronal scaffold protein and the implications it may have for memory and cognition. Finally, we discuss the evidence supporting a role for LRP in AD. Although the evidence for LRP as a genetic risk factor is weak, many of its ligands impose genetic risk, and have been implicated in AD pathogenic cascades. We discuss the role of LRP in amyloid precursor protein (APP) processing and production of beta-amyloid (Abeta. We identify LRP ligands that accelerate aggregation of toxic Abeta species. LRP mediates crucial pathways in AD pathogenesis such as Abeta clearance, Abeta uptake, intraneuronal Abeta accumulation and Abeta-associated neuron death. Interestingly, the TGFbeta -V receptor is LRP-1. Data show that one critical ligand TGFbeta2, associated with neurodegeneration in amyloid diseases, induces LRP expression in PC12 cells. Data from rodent infusion models demonstrate the impact of TGFbeta2 in modifying Abeta- induced Long Term Potentiation (LTP) responses, presynaptic proteins, lipid peroxidation, gliosis and staining for neuronal nuclei. The evidence supports a complex and significant role of LRP in cognition and AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cognition/physiology , LDL-Receptor Related Proteins/metabolism , Neurons/metabolism , Animals , Humans , Ligands , Mice , Mice, Knockout , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology
4.
J Neurochem ; 93(4): 1038-46, 2005 May.
Article in English | MEDLINE | ID: mdl-15857407

ABSTRACT

Apolipoprotein J (apoJ; also known as clusterin and sulfated glycoprotein (SGP)-2) is associated with senile plaques in degenerating regions of Alzheimer's disease brains, where activated microglia are also prominent. We show a functional link between apoJ and activated microglia by demonstrating that exogenous apoJ activates rodent microglia in vivo and in vitro. Intracerebroventricular infusion of purified human plasma apoJ ( approximately 4 microg over 28 days) activated parenchymal microglia to a phenotype characterized by enlarged cell bodies and processes (phosphotyrosine immunostaining). In vitro, primary rat microglia were also activated by apoJ, with changes in morphology and induction of major histocompatibility complex class II (MHCII) antigen. ApoJ increased the secretion of reactive nitrogen intermediates in a dose-dependent manner (EC(50) 112 nm), which was completely blocked by aminoguanidine (AG), a nitric oxide synthase inhibitor. However, AG did not block the increased secretion of tumor necrosis factor-alpha by apoJ (EC(50) 55 nm). Microglial activation by apoJ was also blocked by an anti-apoJ monoclonal antibody (G7), and by chemical cleavage of apoJ with 2-nitro-5-thiocyanobenzoate. The mitogen-activated protein kinase kinase and protein kinase C inhibitors PD98059 and H7 inhibited apoJ-mediated induction of reactive nitrogen intermediate secretion from cultured microglia. As a functional measure, apoJ-activated microglia secreted neurotoxic agents in a microglia-neuron co-culture model. We hypothesize that ApoJ contributes to chronic inflammation and neurotoxicity through direct effects on microglia.


Subject(s)
Cerebral Cortex/cytology , Complement Inactivator Proteins/pharmacology , Glycoproteins/pharmacology , Microglia/drug effects , Molecular Chaperones/pharmacology , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Clusterin , Complement Hemolytic Activity Assay/methods , Complement Inactivator Proteins/isolation & purification , Diagnostic Imaging/methods , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Flavonoids/pharmacology , Glycoproteins/isolation & purification , Humans , Immunohistochemistry/methods , In Vitro Techniques , Interferons/pharmacology , Microglia/metabolism , Molecular Chaperones/isolation & purification , Neurons/drug effects , Neurons/metabolism , Nitrites/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Rats , Rats, Inbred F344 , Thiocyanates/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
5.
Neurochem Int ; 39(5-6): 435-48, 2001.
Article in English | MEDLINE | ID: mdl-11578779

ABSTRACT

The accumulation of fibrillar aggregates of beta Amyloid (A beta) in Alzheimer's Disease (AD) brain is associated with chronic brain inflammation. Although activated microglia (mu glia) can potentially clear toxic amyloid, chronic activation may lead to excessive production of neurotoxins. Recent epidemiological and clinical data have raised questions about the use of anti-inflammatory steroids (glucocorticoids, Gcs) and estrogens for treatment or prevention of AD. Since very little is known about steroid effects on mu glial interactions with amyloid, we investigated the effects of the synthetic Gc dexamethasone (DXM) and 17-beta estradiol (E2) in vitro in a murine mu glial-like N9 cell line on toxin production and intracellular A beta accumulation. To determine whether the steroid alterations of A beta uptake in vitro had relevance in vivo, we examined the effects of these steroids on A beta accumulation and mu glial responses to A beta infused into rat brain. Our in vitro data demonstrate for the first time that Gc dose-dependently enhanced mu glial A beta accumulation and support previous work showing that E2 enhances A beta uptake. Despite both steroids enhancing uptake, degradation was impeded, particularly with Gcs. Distinct differences between the two steroids were observed in their effect on toxin production and cell viability. Gc dose-dependently increased toxicity and potentiated A beta induction of nitric oxide, while E2 promoted cell viability and inhibited A beta induction of nitric oxide. The steroid enhancement of mu glial uptake and impedence of degradation observed in vitro were consistent with observations from in vivo studies. In the brains of A beta-infused rats, the mu glial staining in entorhinal cortex layer 3, not associated with A beta deposits was increased in response to A beta infusion and this effect was blocked by feeding rats prednisolone. In contrast, E2 enhanced mu glial staining in A beta-infused rats. A beta-immunoreactive (ir) deposits were quantitatively smaller, appeared denser, and were associated with robust mu glial responses. Despite the fact that steroid produced a smaller more focal deposit, total extracted A beta in cortical homogenate was elevated. Together, the in vivo and in vitro data support a role for steroids in plaque compaction. Our data are also consistent with the hypothesis that although E2 is less potent than Gc in impeding A beta degradation, long term exposure to both steroids could reduce A beta clearance and clinical utility. These data showing Gc potentiation of A beta-induced mu glial toxins may help explain the lack of epidemiological correlation for AD. The failure of both steroids to accelerate A beta degradation may explain their lack of efficacy for treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Dexamethasone/pharmacology , Estradiol/pharmacology , Glucocorticoids/pharmacology , Microglia/drug effects , Prednisolone/pharmacology , Amyloid beta-Peptides/pharmacology , Animals , Brain/cytology , Brain/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Mice , Microglia/metabolism , Microglia/physiology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Toxins, Biological/biosynthesis
6.
J Neurosci ; 21(21): 8370-7, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11606625

ABSTRACT

Inflammation in Alzheimer's disease (AD) patients is characterized by increased cytokines and activated microglia. Epidemiological studies suggest reduced AD risk associates with long-term use of nonsteroidal anti-inflammatory drugs (NSAIDs). Whereas chronic ibuprofen suppressed inflammation and plaque-related pathology in an Alzheimer transgenic APPSw mouse model (Tg2576), excessive use of NSAIDs targeting cyclooxygenase I can cause gastrointestinal, liver, and renal toxicity. One alternative NSAID is curcumin, derived from the curry spice turmeric. Curcumin has an extensive history as a food additive and herbal medicine in India and is also a potent polyphenolic antioxidant. To evaluate whether it could affect Alzheimer-like pathology in the APPSw mice, we tested a low (160 ppm) and a high dose of dietary curcumin (5000 ppm) on inflammation, oxidative damage, and plaque pathology. Low and high doses of curcumin significantly lowered oxidized proteins and interleukin-1beta, a proinflammatory cytokine elevated in the brains of these mice. With low-dose but not high-dose curcumin treatment, the astrocytic marker GFAP was reduced, and insoluble beta-amyloid (Abeta), soluble Abeta, and plaque burden were significantly decreased by 43-50%. However, levels of amyloid precursor (APP) in the membrane fraction were not reduced. Microgliosis was also suppressed in neuronal layers but not adjacent to plaques. In view of its efficacy and apparent low toxicity, this Indian spice component shows promise for the prevention of Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid/metabolism , Antioxidants/administration & dosage , Curcumin/administration & dosage , Oxidative Stress/drug effects , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid/drug effects , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalitis/complications , Encephalitis/drug therapy , Encephalitis/pathology , Enzyme Inhibitors/administration & dosage , Female , Glial Fibrillary Acidic Protein/metabolism , Interleukin-1/metabolism , Male , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Oxidation-Reduction/drug effects , Solubility/drug effects , Spices
8.
Neurobiol Aging ; 22(6): 983-91, 2001.
Article in English | MEDLINE | ID: mdl-11755007

ABSTRACT

We previously showed the non-steroidal anti-inflammatory drug (NSAID) ibuprofen suppresses inflammation and amyloid in the APPsw (Tg2576) Tg2576 transgenic mouse. The mechanism for these effects and the impact on behavior are unknown. We now show ibuprofen's effects were not mediated by alterations in amyloid precursor protein (APP) expression or oxidative damage (carbonyls). Six months ibuprofen treatment in Tg+ females caused a decrease in open field behavior (p < 0.05), restoring values similar to Tg- mice. Reduced caspase activation per plaque provided further evidence for a neuroprotective action of ibuprofen. The impact of a shorter 3 month duration ibuprofen trial, beginning at a later age (from 14 to 17 months), was also investigated. Repeated measures ANOVA of Abeta levels (soluble and insoluble) demonstrated a significant ibuprofen treatment effect (p < 0.05). Post-hoc analysis showed that ibuprofen-dependent reductions of both soluble Abeta and Abeta42 were most marked in entorhinal cortex (p < 0.05). Although interleukin-1beta and insoluble Abeta were more effectively reduced with longer treatment, the magnitude of the effect on soluble Abeta was not dependent on treatment duration.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Behavior, Animal/drug effects , Ibuprofen/pharmacology , Aging/pathology , Aging/psychology , Alzheimer Disease/genetics , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Interleukin-1/metabolism , Male , Mice , Mice, Transgenic , Oxidation-Reduction , Sex Characteristics
9.
Neurobiol Aging ; 22(6): 993-1005, 2001.
Article in English | MEDLINE | ID: mdl-11755008

ABSTRACT

Both oxidative damage and inflammation have been implicated in age-related neurodegenerative diseases including Alzheimer's Disease (AD). The yellow curry spice, curcumin, has both antioxidant and anti-inflammatory activities which confer significant protection against neurotoxic and genotoxic agents. We used 22 month Sprague-Dawley (SD) rats to compare the effects of the conventional NSAID, ibuprofen, and curcumin for their ability to protect against amyloid beta-protein (Abeta)-induced damage. Lipoprotein carrier-mediated, intracerebroventricular infusion of Abeta peptides induced oxidative damage, synaptophysin loss, a microglial response and widespread Abeta deposits. Dietary curcumin (2000 ppm), but not ibuprofen, suppressed oxidative damage (isoprostane levels) and synaptophysin loss. Both ibuprofen and curcumin reduced microgliosis in cortical layers, but curcumin increased microglial labeling within and adjacent to Abeta-ir deposits. In a second group of middle-aged female SD rats, 500 ppm dietary curcumin prevented Abeta-infusion induced spatial memory deficits in the Morris Water Maze and post-synaptic density (PSD)-95 loss and reduced Abeta deposits. Because of its low side-effect profile and long history of safe use, curcumin may find clinical application for AD prevention.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Cognition Disorders/chemically induced , Neurotoxins/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Phenols/pharmacology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/toxicity , Brain , Brain Chemistry , Cognition Disorders/pathology , Cognition Disorders/psychology , Curcumin/pharmacology , Diet , Enzyme-Linked Immunosorbent Assay , Ibuprofen/pharmacology , Image Interpretation, Computer-Assisted , Immunohistochemistry , Injections , Maze Learning/drug effects , Molecular Chaperones/chemistry , Neurotoxins/toxicity , Oxidation-Reduction , Peptide Fragments/administration & dosage , Peptide Fragments/toxicity , Plaque, Amyloid/pathology , Synaptophysin/metabolism
10.
J Neurosci ; 20(15): 5709-14, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10908610

ABSTRACT

The brain in Alzheimer's disease (AD) shows a chronic inflammatory response characterized by activated glial cells and increased expression of cytokines and complement factors surrounding amyloid deposits. Several epidemiological studies have demonstrated a reduced risk for AD in patients using nonsteroidal anti-inflammatory drugs (NSAIDs), prompting further inquiries about how NSAIDs might influence the development of AD pathology and inflammation in the CNS. We tested the impact of chronic orally administered ibuprofen, the most commonly used NSAID, in a transgenic model of AD displaying widespread microglial activation, age-related amyloid deposits, and dystrophic neurites. These mice were created by overexpressing a variant of the amyloid precursor protein found in familial AD. Transgene-positive (Tg+) and negative (Tg-) mice began receiving chow containing 375 ppm ibuprofen at 10 months of age, when amyloid plaques first appear, and were fed continuously for 6 months. This treatment produced significant reductions in final interleukin-1beta and glial fibrillary acidic protein levels, as well as a significant diminution in the ultimate number and total area of beta-amyloid deposits. Reductions in amyloid deposition were supported by ELISA measurements showing significantly decreased SDS-insoluble Abeta. Ibuprofen also decreased the numbers of ubiquitin-labeled dystrophic neurites and the percentage area per plaque of anti-phosphotyrosine-labeled microglia. Thus, the anti-inflammatory drug ibuprofen, which has been associated with reduced AD risk in human epidemiological studies, can significantly delay some forms of AD pathology, including amyloid deposition, when administered early in the disease course of a transgenic mouse model of AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ibuprofen/pharmacology , Plaque, Amyloid/pathology , Alzheimer Disease/immunology , Amyloidosis/drug therapy , Amyloidosis/immunology , Amyloidosis/pathology , Animals , Brain/immunology , Brain/pathology , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Interleukin-1/metabolism , Male , Mice , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Neuroimmunomodulation/drug effects , Plaque, Amyloid/immunology , Ubiquitins/metabolism
11.
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
12.
Neuroreport ; 10(14): 2911-7, 1999 Sep 29.
Article in English | MEDLINE | ID: mdl-10549796

ABSTRACT

We evaluated the relationship between amyloid-beta protein (A beta) concentration and the metabolic abnormality in an Alzheimer's disease (AD) patient as measured by [18F]fluorodeoxyglucose positron emission tomography (FDG-PET). Across most regions there were significant inverse correlations among FDG-PET intensity values and both insoluble. The temporal lobe samples showed no significant correlation between FDG-PET values and A beta deposition. Findings support A beta as contributing to the hypometabolism in regions of the AD brain that are still relatively viable metabolically; those regions with chronic pathologic damage, such as temporal cortex, may have other factors that contribute to metabolic deficits.


Subject(s)
Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Aged , Aged, 80 and over , Algorithms , Body Burden , Brain Chemistry/physiology , Brain Mapping , Fluorodeoxyglucose F18 , Humans , Image Processing, Computer-Assisted , Male , Radiopharmaceuticals , Tomography, Emission-Computed
13.
J Neurosci Res ; 57(4): 504-20, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10440900

ABSTRACT

An inflammatory response involving activated microglia in neuritic beta-amyloid plaques is found in Alzheimer's disease (AD) brain. Because HDL lipoproteins have been shown to carry the beta-amyloid peptide (Abeta) in plasma and CSF, we have investigated the influence of plasma high-density lipoprotein (HDL) and lipidated ApoE and ApoJ particles on the interaction of cultured rat microglia with Abeta1-42. Microglia degraded Abeta via a pathway sensitive to cytochalasin D and the scavenger receptor inhibitor, fucoidan. HDL increased the degradation of Abeta and the ratio of multimeric/monomeric Abeta in a dose-dependent manner. In contrast, lipidated ApoJ and ApoE decreased the degradation of Abeta, and the effects were ApoE isoform-dependent. Immuno-electron microscopy revealed internalized Abeta in endosomes and lysosomes as well as cell-associated Abeta in deep invaginations, which may be related to caveolae and surface-connected compartments. These data suggest that lipoprotein-dependent Abeta trafficking to microglia could be relevant to plaque pathogenesis in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Lipoproteins/pharmacology , Microglia/drug effects , Plaque, Amyloid/metabolism , Animals , Blotting, Western , Cell Count/drug effects , Cells, Cultured , Humans , Immunohistochemistry , Microglia/metabolism , Microscopy, Electron , Rats
14.
Neuroscience ; 91(3): 1009-16, 1999.
Article in English | MEDLINE | ID: mdl-10391478

ABSTRACT

A role for apolipoprotein E is implicated in regeneration of synaptic circuitry after neural injury. The in vitro mouse organotypic hippocampal slice culture system shows Timm's stained mossy fiber sprouting into the dentate gyrus molecular layer in response to deafferentation of the entorhinal cortex. We show that cultures derived from apolipoprotein E knockout mice are defective in this sprouting response; specifically, they show no sprouting in the dorsal region of the dentate gyrus, yet retain sprouting in the ventral region. Dorsal but not ventral sprouting in cultures from C57B1/6J mice is increased 75% by treatment with 100 pM 17beta-estradiol; this response is blocked by both progesterone and tamoxifen. These results show that neuronal sprouting is increased by estrogen in the same region where sprouting is dependent on apolipoprotein E. Sprouting may be stimulated by estrogen through its up-regulation of apolipoprotein E expression leading to increased recycling of membrane lipids for use by sprouting neurons. Estrogen and apolipoprotein E may therefore interact in their modulation of both Alzheimer's disease risk and recovery from CNS injury.


Subject(s)
Apolipoproteins E/physiology , Estradiol/pharmacology , Mossy Fibers, Hippocampal/drug effects , Mossy Fibers, Hippocampal/physiology , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Hippocampus/metabolism , Immunologic Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Organ Culture Techniques
15.
J Neurosci ; 18(24): 10366-74, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9852574

ABSTRACT

The transforming growth factor-beta (TGF-beta) family consists of three isoforms and is part of a larger family of cytokines regulating differentiation, development, and tissue repair. Previous work from our laboratory has shown that TGF-beta1 can increase amyloid-beta protein (Abeta) immunoreactive (Abetair) plaque-like deposits in rat brain. The aim of the current study was to evaluate all three isoforms of TGF-beta for their ability to affect the deposition and neurotoxicity of Abeta in an organotypic, hippocampal slice culture model of Abeta deposition. Slice cultures were treated with Abeta either with or without one of the TGF-beta isoforms. All three isoforms can increase Abeta accumulation (over Abeta treatment alone) within the slice culture, as determined by ELISA. However, there are striking differences in the pattern of Abetair among the three isoforms of TGF-beta. Isoforms 1 and 3 produced a cellular pattern of Abeta staining that colocalizes with GS lectin staining (microglia). TGF-beta2 produces dramatic Abeta staining of pyramidal neurons in layers CA1-CA2. In addition to cellular Abeta staining, plaque-like deposits are increased by all of the TGF-betas. Although no gross toxicity was observed, morphological neurodegenerative changes were seen in the CA1 region when the slices were treated with Abeta plus TGF-beta2. Our results demonstrate important functional differences among the TGF-beta isoforms in their ability to alter the cellular distribution and degradation of Abeta. These changes may be relevant to the pathology of Alzheimer's disease (AD).


Subject(s)
Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Antibodies/pharmacology , Culture Media, Conditioned/chemistry , Enzyme-Linked Immunosorbent Assay , Hippocampus/drug effects , Hippocampus/pathology , Immunohistochemistry , In Vitro Techniques , Inflammation/metabolism , Mice , Mice, Inbred ICR , Nerve Degeneration/pathology , Protein Isoforms/pharmacology , Time Factors , Tumor Necrosis Factor-alpha/analysis
16.
FEBS Lett ; 436(3): 439-44, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9801165

ABSTRACT

Murine N9 microglia accumulated A beta from media containing 0.67 microM A beta within 6 h. In N9 and in primary rat microglia, chloroquine, which disrupts lysosomal pH, increased A beta-induced accumulation of A beta, particularly A beta1-42. Leupeptin similarly enhanced A beta accumulation. The scavenger receptor antagonist fucoidan did not affect acute chloroquine-dependent A beta1-42 accumulation, demonstrating uptake of non-aggregated A beta. After prolonged incubations, chloroquine enhanced A beta multimer (8-12 kDa) accumulation, an effect inhibited by fucoidan. Disruptions of the lysosomal system enhance A beta and its multimer formation. Despite negligible effects of fucoidan on initial A beta uptake, chronic exposure inhibits multimer accumulation, demonstrating a role for scavenger receptor in multimer accumulation.


Subject(s)
Amyloid beta-Peptides/pharmacokinetics , Chloroquine/pharmacology , Leupeptins/pharmacology , Membrane Proteins , Microglia/metabolism , Peptide Fragments/pharmacokinetics , Receptors, Lipoprotein , Animals , Biological Transport/drug effects , Cell Line , Cells, Cultured , Kinetics , Mice , Microglia/cytology , Microglia/drug effects , Polysaccharides/pharmacology , Rats , Receptors, Immunologic/antagonists & inhibitors , Receptors, Scavenger , Scavenger Receptors, Class B
17.
J Neurosci ; 18(20): 8311-21, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9763475

ABSTRACT

Amyloid beta-protein, Abeta, is normally produced in brain and is cleared by unknown mechanisms. In Alzheimer's disease (AD), Abeta accumulates in plaque-like deposits and is implicated genetically in neurodegeneration. Here we investigate mechanisms for Abeta degradation and Abeta toxicity in vivo, focusing on the effects of Abeta40, which is the peptide that accumulates in apolipoprotein E4-associated AD. Chronic intraventricular infusion of Abeta40 into rat brain resulted in limited deposition and toxicity. Coinfusion of Abeta40 with the cysteine protease inhibitor leupeptin resulted in increased extracellular and intracellular Abeta immunoreactivity. Analysis of gliosis and TUNEL in neuron layers of the frontal and entorhinal cortex suggested that leupeptin exacerbated Abeta40 toxicity. This was supported further by the neuronal staining of cathepsin B in endosomes or lysosomes, colocalizing with intracellular Abeta immunoreactivity in pyknotic cells. Leupeptin plus Abeta40 caused limited but significant neuronal phospho-tau immunostaining in the entorhinal cortex. Intriguingly, Abeta40 plus leupeptin induced intracellular accumulation of the more toxic Abeta, Abeta42, in a small group of septal neurons. Leupeptin infusion previously has been reported to interfere with lysosomal proteolysis and to result in the accumulation of lipofuscin, dystrophic neurites, tau- and ubiquitin-positive inclusions, and structures resembling paired helical filaments. Coinfusion of Abeta40 with the serine protease inhibitor aprotinin also increased diffuse extracellular deposition but reduced astrocytosis and TUNEL and was not associated with intracellular Abeta staining. Collectively, these data suggest that an age or Alzheimer's-related defect in lysosomal/endosomal function could promote Abeta deposition and DNA fragmentation in neurons and glia similar to that found in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloidosis/metabolism , Neurons/pathology , Protease Inhibitors/pharmacology , Alzheimer Disease/pathology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/immunology , Amyloidosis/pathology , Animals , Antibodies, Monoclonal , Aprotinin/pharmacology , Cathepsins/metabolism , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Glial Fibrillary Acidic Protein/analysis , Gliosis/metabolism , In Situ Nick-End Labeling , Leupeptins/pharmacology , Lysosomes/chemistry , Lysosomes/physiology , Microscopy, Confocal , Neuroglia/chemistry , Neuroglia/physiology , Neurons/enzymology , Neurotoxins/metabolism , Rats , Rats, Sprague-Dawley , Serine Proteinase Inhibitors/pharmacology , Thalamus/metabolism , Thalamus/pathology
18.
J Neurophysiol ; 79(2): 1045-52, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9463460

ABSTRACT

Complex patterns of intercellular calcium signaling occur in the CA1 and CA2 regions of hippocampal slice organotypic cultures from neonatal mice. Spontaneous localized intercellular Ca2+ waves involving 5-15 cells propagate concentrically from multiple foci in the stratum oriens and s. radiatum. In these same regions, extensive Ca2+ waves involving hundreds of cells propagate as curvilinear and spiral wavefronts across broad areas of CA1 and CA2. Ca2+ waves travel at rates of 5-10 mu m/s, are abolished by thapsigargin, and do not require extracellular Ca2+. Staining for astrocytes and neurons indicates that these intercellular waves occur primarily in astrocytes. The frequency and amplitude of Ca2+ waves increase in response to bath application of N-methyl-D-aspartate (NMDA) and decrease in response to removal of extracellular Ca2+ or application of tetrodotoxin. This novel pattern of intercellular Ca2+ signaling is characteristic of the behavior of an excitable medium. Networks of glial cells in the hippocampus may behave as an excitable medium whose spatial and temporal signaling properties are modulated by neuronal activity.


Subject(s)
Calcium/metabolism , Cell Communication/physiology , Hippocampus/metabolism , Second Messenger Systems/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Cell Communication/drug effects , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Ion Transport/drug effects , Membrane Potentials/drug effects , Mice , N-Methylaspartate/pharmacology , Organ Culture Techniques , Organ Specificity , Oxygen/metabolism , Tetrodotoxin/pharmacology , Thapsigargin/pharmacology
19.
Am J Pathol ; 152(2): 379-89, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9466564

ABSTRACT

During apoptosis, activation of a family of cysteine proteases related to interleukin-1beta-converting enzyme (ICE)-related proteases or "caspases" results in endoproteolytic cleavage of multiple substrates at specific aspartate residues. We have sought to develop new antibody probes for the neoepitopes in protein fragments produced by ICE-related proteolytic cleavage as specific markers of events tightly linked to apoptotic mechanisms. Here, we demonstrate that an antibody probe specific for the C terminus of a 32-kd actin fragment produced by ICE-like activity specifically labels apoptotic but not necrotic, differentiated human neuroblastoma cells in culture. Unlike probes for nonspecific DNA strand breaks confined to the nucleus or cell body, this method allows the detection of cytoskeletal fragments in cell processes as well as the perikaryon long before DNA fragmentation and cell death and therefore serves as a novel marker of apoptosis-related events in distal parts of cells such as axons and dendrites. To illustrate this new tool, we show that the antibody detects the processes and cell bodies of degenerating neurons and plaque-associated microglia in Alzheimer's disease. In situ detection of caspase-cleaved actin provides a new means to evaluate the role of caspase activation in pathological and physiological processes.


Subject(s)
Actins/immunology , Alzheimer Disease/pathology , Antibodies/immunology , Apoptosis/physiology , Neuroblastoma/pathology , Peptide Fragments/immunology , Actins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Brain/metabolism , Brain/pathology , Caspase 1 , Cysteine Endopeptidases/metabolism , Endopeptidases/metabolism , Humans , Immunohistochemistry/methods , Microglia/pathology , Middle Aged , Neuroblastoma/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Plaque, Amyloid/pathology , Tumor Cells, Cultured
20.
Am J Pathol ; 152(1): 307-17, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9422548

ABSTRACT

Microglial activation is central to the inflammatory response in Alzheimer's Disease (AD). A recently described mouse line, Tg(HuAPP695.K670N/M671L)2576, expressing human amyloid precursor protein with a familial AD gene mutation, age-related amyloid deposits, and memory deficits, was found to develop a significant microglial response using Griffonia simplicifolia lectin or phosphotyrosine probe to identify microglia Both Griffonia simplicifolia lectin and phosphotyrosine staining showed increased numbers of intensely labeled, often enlarged microglia clustered in and around plaques, consistent with microglial activation related to beta-amyloid formation. Using quantitative image analysis of coronal phosphotyrosine-immunostained sections, transgene-positive 10- to 16-month-old, hemizygous, hybrid Tg2576 (APPsw) animals showed significantly increased microglial density and size in plaque-forming areas of hippocampus and frontal, entorhinal, and occipital cortex. Quantitative analysis of microglia as a function of distance from the center of plaques (double labeled for A beta peptide and microglia) revealed highly significant, two- to fivefold elevations in microglial number and area within plaques compared with neighboring regions. Tg2576 beta-amyloid-plaque-forming mice should be a useful system for assessing the consequences of the microglial-mediated inflammatory response to beta-amyloid and developing anti-inflammatory therapeutic strategies for Alzheimer's disease. These results provide the first quantitative link between beta-amyloid plaque formation and microglial activation in an animal model with neuritic plaques and memory deficits.


Subject(s)
Mice, Transgenic/physiology , Microglia/physiology , Plaque, Amyloid/physiology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Cell Count , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Microglia/pathology , Mutation , Plaque, Amyloid/pathology , Transgenes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...