Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
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
2.
Neurology ; 71(2): 85-92, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18509095

ABSTRACT

BACKGROUND: Persons at risk for familial Alzheimer disease (FAD) provide a model in which biomarkers can be studied in presymptomatic disease. METHODS: Twenty-one subjects at risk for presenilin-1 (n = 17) or amyloid precursor protein (n = 4) mutations underwent evaluation with the Clinical Dementia Rating (CDR) scale. We obtained plasma from all subjects and CSF from 11. Plasma (Abeta(40), Abeta(42), F(2)-isoprostanes) and CSF (F(2)-isoprostanes, t-tau, p-tau(181), Abeta(40), Abeta(42), and Abeta(42)/Abeta(40) ratio) levels were compared between FAD mutation carriers (MCs) and noncarriers (NCs). RESULTS: Plasma Abeta(42) levels (25.1 pM vs 15.5 pM, p = 0.031) and the ratio of Abeta(42)/Abeta(40) (0.16 vs 0.11, p = 0.045) were higher in presymptomatic MCs. Among MCs, those with CDR scores of 0.5 had lower plasma Abeta(42) levels than those with CDR scores of 0 (14.1 pM vs 25.1, p = 0.02). The ratio of Abeta(42) to Abeta(40) was also reduced in the CSF (0.08 vs 0.15, p = 0.046) of nondemented MCs compared to NCs. Total CSF tau and p-tau(181) levels were elevated in presymptomatic FAD MCs. CSF levels of F(2)-isoprostanes were also elevated in MCs (n = 7, 48.6 pg/mL) compared to NCs (n = 4, 21.6 pg/mL, p = 0.031). CONCLUSIONS: Our data indicate that Abeta(42) is elevated in plasma in familial Alzheimer disease (FAD) mutation carriers (MCs) and suggests that this level may decrease with disease progression prior to the development of overt dementia. We also demonstrated that the ratio of Abeta(42) to Abeta(40) was reduced in the CSF of nondemented MCs and that elevations of t-tau and p-tau(181) are sensitive indicators of presymptomatic disease. Our finding of elevated F(2)-isoprostane levels in the CSF of preclinical FAD MCs suggests that oxidative stress occurs downstream to mismetabolism of amyloid precursor protein.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/prevention & control , Heterozygote , Adult , Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Female , Humans , Isoprostanes/blood , Isoprostanes/cerebrospinal fluid , Male , Mutation , Neurologic Examination , Presenilin-1/genetics , Protease Nexins , Receptors, Cell Surface/genetics , Sensitivity and Specificity , tau Proteins/cerebrospinal fluid
3.
Neuroscience ; 117(3): 723-30, 2003.
Article in English | MEDLINE | ID: mdl-12617976

ABSTRACT

Epidemiological studies have suggested that the chronic use of non-steroidal anti-inflammatory drugs (NSAIDs) reduces the relative risk of Alzheimer's disease (AD). The possible neuroprotection by NSAIDs in AD is generally attributed to anti-inflammatory activity. An additional mode of drug action may involve anti-aggregation of beta-amyloid (Abeta) peptides by commonly used NSAIDs. We utilized in vitro competition assays, autoradiography, and fluorescence microscopy with AD brain specimens to demonstrate concentration-dependent decreases in the binding of the in vivo molecular imaging probe, 2-(1-[6-[(2-[(18)F]fluoroethyl)(methyl)amino]-2-naphthyl]ethylidene)malononitrile ([(18)F]FDDNP), against (S)-naproxen and (R)- and (S)-ibuprofen (but not diclofenac) to Abeta fibrils and ex vivo Abeta senile plaques. Conversely, in vitro amyloid dyes Congo Red and Thioflavine T were demonstrated in the same experiments not to bind to the FDDNP binding site. FDDNP and the NSAIDs that share the same binding site also exhibit anti-aggregation effects on Abeta peptides, suggesting that the shared binding site on Abeta fibrils and plaques may be a site of anti-aggregation drug action. Our results indicate for the first time the binding of select NSAIDs to plaques, specifically to the binding site of the molecular imaging probe [(18)F]FDDNP. Our understanding of the molecular requirements of FDDNP binding may help in the optimization of the Abeta anti-aggregation potency of experimental drugs. [(18)F]FDDNP has been used to image plaques in vivo with positron emission tomography (PET), and investigations into the influence of Abeta anti-aggregation on the risk-reduction effects of NSAIDs on AD could utilize [(18)F]FDDNP and PET in determining the occupancy rate of NSAIDs and experimental drugs in plaques in the living brain of AD patients.


Subject(s)
Alzheimer Disease/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Ibuprofen/pharmacokinetics , Naproxen/pharmacokinetics , Tomography, Emission-Computed/methods , Aged , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Autoradiography/methods , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive , Brain/metabolism , Brain/physiopathology , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Nitriles , Plaque, Amyloid/diagnostic imaging , Radiopharmaceuticals
4.
J Neurochem ; 83(4): 1009-12, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12421374

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) have been associated with reduced risk for Alzheimer's disease (AD) and selected NSAIDs racemates suppress beta-amyloid (Abeta) accumulation in vivo and Abeta42 production in vitro. Clinical use of NSAIDs for preventing or treating AD has been hampered by dose-limiting toxicity believed to be due to cyclooxygenase (COX)-inhibition that is reportedly not essential for selective Abeta42 reduction. Profens have racemates and R-enantiomers were supposed to be inactive forms. Here we demonstrate that R-ibuprofen and R-flurbiprofen, with poor COX-inhibiting activity, reduce Abeta42 production by human cells. Although these R-enantiomers inhibit nuclear factor-kappaB (NF-kappaB) activation and NF-kappaB can selectively regulate Abeta42, Abeta42 reduction is not mediated by inhibition of NF-kappaB activation. Because of its efficacy at lowering Abeta42 production and low toxicity profile, R-flurbiprofen is a strong candidate for clinical development.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Amyloid beta-Peptides/analysis , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Blotting, Western , Cell Line , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Dose-Response Relationship, Drug , Flurbiprofen/pharmacology , Humans , Ibuprofen/pharmacology , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Mutation , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Peptide Fragments/analysis , Peptides/pharmacology , Stereoisomerism , Transfection
5.
J Neurosci Res ; 68(3): 331-6, 2002 May 01.
Article in English | MEDLINE | ID: mdl-12111863

ABSTRACT

The apolipoprotein E (apoE) epsilon 4 allele (apoE4) is a major risk factor for neurodegenerative conditions, including Alzheimer's disease (AD). A role for apoE in regeneration of synaptic circuitry after neural injury has been shown in several in vitro studies in which apoE3 supports neuronal sprouting better than apoE4. We evaluated sprouting in an in vitro mouse organotypic hippocampal slice culture system derived from transgenic mice expressing apoE3 or apoE4, in which apoE-dependent granule cell mossy fiber sprouting in the presence of apoE4 is only 51% of the level of apoE3. Sprouting supported by apoE4 had a dose response opposite that by supported by apoE3: although increasing E3 expression increased sprouting, increasing E4 expression decreased sprouting, suggesting that the defect in E4 in supporting neuronal sprouting is a gain-of-negative activity. These results may have important pharmacogenomic implications for AD therapies that modulate apoE expression levels.


Subject(s)
Alzheimer Disease/genetics , Apolipoproteins E/metabolism , Cell Differentiation/genetics , Growth Cones/metabolism , Mossy Fibers, Hippocampal/growth & development , Neuronal Plasticity/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Animals, Newborn , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/genetics , Female , Gene Expression Regulation, Developmental/physiology , Genetic Predisposition to Disease/genetics , Growth Cones/ultrastructure , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mossy Fibers, Hippocampal/metabolism , Mossy Fibers, Hippocampal/ultrastructure , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , Up-Regulation/genetics
6.
Acta Neuropathol ; 103(1): 66-73, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11841033

ABSTRACT

Human perinatal hypoxic-ischemic brain injury is an important cause of death and morbidity. One relatively common pattern of perinatal injury involves selective neuronal death in the ventral gray matter of the pons and in the subiculum of the hippocampal formation, classically termed 'pontosubicular neuronal necrosis' (PSN). The vulnerable neurons undergo karyorrhectic condensation of their nuclear chromatin and exhibit in situ end labeling for DNA fragmentation, leading to the recent reclassification of cell death in PSN as apoptotic. Caspase activation plays a central role in apoptosis and caspase-3 appears to be an especially important effector enzyme in neuronal apoptosis. In this study we performed immunohistochemistry on brain sections from six postmortem cases of PSN using two polyclonal antisera; CM1, a specific marker of caspase-3 activation, and fractin, which specifically recognizes a neoepitope at a caspase cleavage site in actin, and is therefore a marker of caspase-like proteolytic activity. Numerous CM1- and fractin-immunolabeled neurons were seen in the nuclei pontis and subiculum in each case, and the great majority showed karyorrhectic morphology. The immunostaining involved the nuclei and cytoplasm of these cells and the proximal portions at least of their neuritic processes. Some neurons exhibited a more extensive pattern of dendritic fractin labeling. Frequent CM1- and fractin-immunoreactive axonal segments were also seen. The identification of caspase-3 activation and caspase-like proteolytic activity in PSN cases in this study suggests that caspase inhibitors may potentially have a therapeutic neuroprotective role in human perinatal hypoxic-ischemic brain injury.


Subject(s)
Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Caspases/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Actins/analysis , Actins/metabolism , Apoptosis , Caspase 3 , Female , Humans , Immunohistochemistry , Infant, Newborn , Male , Neurons/chemistry , Neurons/enzymology , Neurons/pathology , Pons/chemistry , Pons/enzymology , Pons/pathology
7.
J Neurosci ; 21(24): RC189, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11734604

ABSTRACT

Senile plaques (SPs) and neurofibrillary tangles (NFTs) are hallmark pathologies accompanying the neurodegeneration involved in Alzheimer's disease (AD), for which beta-amyloid (Abeta) peptide is a major constituent of SPs. Our laboratories previously developed the hydrophobic, fluorescent molecular-imaging probe 2-(1-(6-[(2-[(18)F]fluoroethyl)(methyl)amino]-2-naphthyl)ethylidene)malononitrile ([(18)F]FDDNP), which crosses the blood-brain barrier and determines the localization and load of SPs and NFTs in vivo in AD patients. In this report, we used fluorimetric and radioactive binding assays to determine the binding affinities of FDDNP and its analog, 1-(6-[(2-[(18)F]fluoroethyl)(methyl)amino]naphthalen-2-yl)ethanone ([(18)F]FENE), to synthetic fibrils of Abeta(1-40). FDDNP and FENE both appeared to bind to two kinetically distinguishable binding sites on Abeta(1-40) fibrils. Fluorescence titrations yielded apparent K(d) values of 0.12 and 0.16 nm for high-affinity binding sites for FDDNP and FENE, respectively, and apparent K(d) values of 1.86 and 71.2 nm for the low-affinity binding sites. The traditional radioactive binding assays also produced apparent K(d) values in the low nanomolar range. The presence of two kinetically distinguishable binding sites for FDDNP and FENE suggests multiple binding sites for SPs and identifies the parameters that allow for the structural optimization of this family of probes for in vivo use. The high-affinity binding of the probes to multiple binding sites on fibrils are consistent with results obtained with digital autoradiography, immunohistochemistry, and confocal fluorescence microscopy using human brain specimens of AD patients.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/metabolism , Naphthalenes/chemistry , Tomography, Emission-Computed , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Autoradiography , Binding Sites/drug effects , Binding Sites/physiology , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Fluorescent Dyes/pharmacokinetics , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Microscopy, Fluorescence , Naphthalenes/pharmacokinetics , Neurofibrillary Tangles/diagnostic imaging , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Nitriles/chemistry , Nitriles/pharmacokinetics , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Plaque, Amyloid/diagnostic imaging , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Radioligand Assay , Substrate Specificity , Tomography, Emission-Computed/methods
8.
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
9.
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
10.
Brain Res ; 904(1): 177-88, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11516428

ABSTRACT

Retinal cell death induced by over-stimulation of glutamate receptors is related to the programmed cell death or apoptosis. However, little is known about the intracellular events that lead to this cell death process in the retina. In this study, we asked if caspase-3 family cysteine proteases regulate cell death in an explant culture of adult rat retina after exposure to excessive glutamate. Cells with DNA fragmentation were first detected in the ganglion cell layer 3 h after a brief exposure to 20 mM glutamate; whilst those in the inner nuclear layer were first observed 6 h after the glutamate lesion. Caspase-3-like activity, as indicated by immunostaining of the fractin antibody that recognizes actin fragments generated by caspase-3 family proteases, was seen 40 min after glutamate treatment. Staining was first detected in the ganglion cell layer and then in the inner nuclear layer, preceding the appearance of cells with DNA fragmentation in these layers. Colocalization study showed that all cells with DNA breaks were fractin positive, indicating that caspase-3 family activity was involved in the glutamate-induced cell death in the adult rat retina. Furthermore, DEVD-CHO, a tetrapeptide inhibitor for caspase-3 family members, reduced dramatically the fractin staining and significantly alleviated glutamate-induced cell death and DNA fragmentation in the ganglion cell layer and inner nuclear layer. Inhibitor for caspase-1-like activity, YVAD-CHO, neither reduced the fractin staining nor showed comparable neuroprotective effects to the retina. We conclude that glutamate-induced apoptotic cell death in adult rat retina is mediated by a specific activation of cysteine proteases related to the caspase-3 family, and an intervention to the caspase-3 proteases provides effective protection to retinal neurons against glutamate excitotoxicity.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Glutamic Acid/pharmacology , Retina/drug effects , Animals , Apoptosis/physiology , Carbocyanines/pharmacokinetics , Caspase 1/metabolism , Caspase 3 , Caspases/metabolism , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation/drug effects , DNA Fragmentation/physiology , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Fluorescent Dyes/pharmacokinetics , Glutamic Acid/metabolism , In Situ Nick-End Labeling , Nerve Degeneration/chemically induced , Nerve Degeneration/enzymology , Nerve Degeneration/physiopathology , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Retina/enzymology , Retina/physiopathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/enzymology , Retinal Ganglion Cells/pathology
11.
Brain Res Brain Res Protoc ; 7(3): 193-202, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11431120

ABSTRACT

A prominent feature of neurodegenerative diseases is a loss of specific neuronal populations. The pathophysiological mechanisms responsible are, however, poorly understood. Primary cultures of rodent embryonic neurons represent a useful experimental system for investigation of molecular pathways of neurodegeneration and mechanisms of cell death. Here, we report a technique utilizing triple-label immunocytochemistry with confocal immunofluorescence detection designed to simultaneously assess multiple parameters of cell injury in individual hippocampal neurons in primary culture. This method combines detection of DNA damage (TUNEL or Klenow assay) with double-label immunocytochemistry for the activated form of caspase-3 or, alternatively, caspase-cleaved actin (fractin), and microtubule-associated protein-2 (MAP-2) or beta-tubulin. The combined evaluation of the form of nuclear damage (karyorrhexis, pyknosis), the presence or absence of activated caspase-3, and the extent of the damage to cell cytoskeleton, allows for precise assessment of the extent of injury and the mode of cell death (apoptosis, oncosis) for individual neurons.


Subject(s)
Immunohistochemistry/methods , Nerve Degeneration/pathology , Neurons/pathology , Animals , Antibodies/chemistry , Antibodies/immunology , Cell Death , Cells, Cultured , Coloring Agents , DNA Damage , Female , Hippocampus/pathology , In Situ Nick-End Labeling , Microscopy, Confocal , Pregnancy , Rats
13.
Mol Cell Neurosci ; 18(5): 503-11, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11922141

ABSTRACT

The amyloid precursor protein presents several cleavage sites leading to the release of its entire C-terminal domain into the cytoplasm. During apoptosis, this C-terminal domain can be cleaved at amino acid 664 by caspases 3, 6, and 8 and can thus generate two peptides N- and C-terminal to amino acid 664 (C31). Recently, it was shown that the C31 induces apoptosis after transfection into N2A and 293 T cell lines. We have analyzed here, by internalization into neurons, the physiological consequences of the entire C-terminal domain (APP-Cter) and of its membrane proximal sequence corresponding to the N-terminal peptide unmasked after caspase cleavage. We find that whereas micromolar concentrations of APP-Cter are harmless, the peptide extending from the membrane (amino acid 649) to the caspase cleavage site (amino acid 664) in the same range of concentrations induces DNA fragmentation, cleavage of actin at a caspase-sensitive site, and activates caspase 3. A mutated version of this sequence (tyrosine 653 replaced by an aspartate) abolishes the effect in vitro and in vivo. Taken together, this report suggests the existence of a new mechanism contributing to Alzheimer's Disease-associated cell death.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Apoptosis/physiology , Brain/metabolism , Cytoplasm/metabolism , Neurons/metabolism , Peptides/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amino Acid Sequence/physiology , Amyloid Precursor Protein Secretases , Animals , Apoptosis/drug effects , Aspartic Acid Endopeptidases , Brain/pathology , Brain/physiopathology , Caspase 3 , Caspases/metabolism , Cells, Cultured , Endopeptidases/metabolism , Female , Male , Mice , Neurons/pathology , Peptide Fragments/metabolism , Peptides/pharmacology , Protein Structure, Tertiary/physiology , Rats
14.
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
15.
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
16.
Neuropathol Appl Neurobiol ; 26(4): 342-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10931367

ABSTRACT

Hirano bodies are eosinophilic rod-like inclusions that are found predominantly in neuronal processes in the hippocampal CA1 sector with increasing age and are particularly numerous in Alzheimer's disease. They contain a variety of cytoskeletal epitopes, especially actin and actin-binding proteins. Actin cleavage by cysteinyl aspartate-specific proteases (caspases) is a feature of apoptosis. Cleavage at aspartate 244 generates N-terminal 32 kDa and C-terminal 15 kDa actin fragments. This has led to the development of a rabbit polyclonal antibody specific for caspase-cleaved actin, directed to the last five C-terminal amino acids of the 32 kDa fragment of actin ('fractin'). Fractin immunohistochemistry was performed on hippocampal sections from Alzheimer's disease and control cases containing numerous Hirano bodies, in addition to immunolabelling with CM1 antiserum which recognizes activated caspase-3. The Hirano bodies showed strong diffuse fractin immunoreactivity. They did not label with CM1 antiserum, perhaps reflecting too low a level of activated caspase-3 for immunodetection, or involvement of a different member of the caspase family. The finding of fractin immunoreactivity of Hirano bodies suggests that caspase-like cleavage of actin may play a role in their formation and further supports caspase-like activity in neuronal processes, distinct from that associated with acute perikaryal apoptosis.


Subject(s)
Actins/metabolism , Alzheimer Disease/metabolism , Caspases/metabolism , Inclusion Bodies/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/pathology , Antibodies/metabolism , Brain Stem/metabolism , Brain Stem/pathology , Caspase 3 , Hippocampus/metabolism , Hippocampus/pathology , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Immune Sera/metabolism , Immunohistochemistry , Inclusion Bodies/ultrastructure
17.
Microsc Res Tech ; 50(4): 316-24, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10936886

ABSTRACT

Amyloid beta-protein (Abeta), the major component of plaques in Alzheimer's disease, is a small hydrophobic protein that is carried on apolipoprotein E (ApoE)- and ApoJ-containing lipoprotein particles in plasma and cerebrospinal fluid (CSF). Microglia, the scavenger cells of the CNS, take up and degrade Abeta via lipoprotein receptors including scavenger receptors A and B, and possibly via other receptors. Lipoproteins, ApoE, and ApoJ influence the uptake and degradation of Abeta in vitro and in vivo. Differences in ApoE-E4, -E3, and -E2 isoforms with respect to Abeta binding to lipoproteins and delivery to cells, including microglia, may contribute to the increased risk of Alzheimer's disease for people with an APOE4 genotype and to risk reduction with APOE2.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Endocytosis , Lipoproteins/metabolism , Microglia/metabolism , Molecular Chaperones , Receptors, Lipoprotein/metabolism , Alzheimer Disease/pathology , Apolipoprotein E2 , Apolipoprotein E4 , Apolipoproteins E/metabolism , Clusterin , Glycoproteins/metabolism , Humans , Microglia/pathology , Microglia/ultrastructure , Protein Isoforms/metabolism
18.
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
19.
J Neurosci Res ; 61(2): 186-92, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10878591

ABSTRACT

Flow cytometry, which definitively identifies each particle as positive or negative with respect to fluorescent markers, is used to characterize the P-2 fraction (crude synaptosomal fraction) with respect to primary components, size, and intactness. Particle size ranged from a few tenths of a microm to greater than 4.5 microm. The viable dye calcein AM labeled 90% of the preparation, indicating that the majority of particles were intact and esterase-positive. 66% of the P-2 fraction is neuronal in origin, as demonstrated by labeling with an antibody directed against SNAP-2. An antibody directed against glial fibrillary acidic protein (GFAP) labeled 35% of the particles in this preparation. The mitochondrial dye nonyl acridine orange (NAO) stained 74% of particles, indicating intra- and extrasynaptosomal mitochondria. Gating analysis reveals that SNAP-25 is enriched in the larger particles. These results suggest that flow cytometry may be used to take advantage of the increased viability, yield, and convenience of the P-2 fraction for studies of nerve terminal function.


Subject(s)
Flow Cytometry , Nerve Tissue Proteins/analysis , Synaptosomes/chemistry , Animals , Coloring Agents , Male , Mitochondria/metabolism , Neuroglia/metabolism , Neuroglia/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley , Scattering, Radiation , Subcellular Fractions/chemistry
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...