Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Mol Psychiatry ; 20(11): 1301-10, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25917367

ABSTRACT

In Alzheimer's disease (AD), neurodegenerative signals such as amyloid-beta (Aß) and the precursors of neurotrophins, outbalance neurotrophic signals, causing synaptic dysfunction and neurodegeneration. The neurotrophin receptor p75 (p75NTR) is a receptor of Aß and mediates Aß-induced neurodegenerative signals. The shedding of its ectodomain from the cell surface is physiologically regulated; however, the function of the diffusible p75NTR ectodomain (p75ECD) after shedding remains largely not known. Here, we show that p75ECD levels in cerebrospinal fluid and in the brains of Alzheimer's patients and amyloid-beta precursor protein (APP)/PS1 transgenic mice were significantly reduced, due to inhibition of the sheddase-tumor necrosis factor-alpha-converting enzyme by Aß. Restoration of p75ECD to the normal level by brain delivery of the gene encoding human p75ECD before or after Aß deposition in the brain of APP/PS1 mice reversed the behavioral deficits and AD-type pathologies, such as Aß deposit, apoptotic events, neuroinflammation, Tau phosphorylation and loss of dendritic spine, neuronal structures and synaptic proteins. Furthermore, p75ECD can also reduce amyloidogenesis by suppressing ß-secretase expression and activities. Our data demonstrate that p75ECD is a physiologically neuroprotective molecule against Aß toxicity and would be a novel therapeutic target and biomarker for AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Brain/pathology , Nerve Tissue Proteins/chemistry , Protein Structure, Tertiary/physiology , Receptors, Nerve Growth Factor/chemistry , ADAM Proteins/metabolism , ADAM17 Protein , Age Factors , Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis/physiology , Brain/drug effects , Brain/metabolism , Case-Control Studies , Cognition Disorders/etiology , Cognition Disorders/therapy , Disease Models, Animal , Down-Regulation/genetics , Humans , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Presenilin-1/genetics , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Recombinant Proteins/therapeutic use , Transduction, Genetic
2.
Neuroscience ; 302: 121-37, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-25286386

ABSTRACT

Multiple cellular systems exist to prevent uncontrolled inflammation in brain tissues; the suppressor of cytokine signaling (SOCS) proteins have key roles in these processes. SOCS proteins are involved in restricting cellular signaling pathways by enhancing the degradation of activated receptors and removing the stimuli for continued activation. There are eight separate SOCS genes that code for proteins with similar structures and properties. All SOCS proteins can reduce signaling of activated transcription factors Janus kinase (JAK) and signal transducer and activator of transcription (STAT), but they also regulate many other signaling pathways. SOCS-1 and SOCS-3 have particular roles in regulating inflammatory processes. Chronic inflammation is a key feature of the pathology present in Alzheimer's disease (AD)-affected brains resulting from responses to amyloid plaques or neurofibrillary tangles, the pathological hallmarks of AD. The goal of this study was to examine SOCS gene expression in human non-demented (ND) and AD brains and in human brain-derived microglia to determine if AD-related pathology resulted in a deficit of these critical molecules. We demonstrated that SOCS-1, SOCS-2, SOCS-3 and cytokine-inducible SH2 containing protein (CIS) mRNA expression was increased in amyloid beta peptide (Aß)- and inflammatory-stimulated microglia, while SOCS-6 mRNA expression was decreased by both types of treatments. Using human brain samples from the temporal cortex from ND and AD cases, SOCS-1 through SOCS-7 and CIS mRNA and SOCS-1 through SOCS-7 protein could be detected constitutively in ND and AD human brain samples. Although, the expression of key SOCS genes did not change to a large extent as a result of AD pathology, there were significantly increased levels of SOCS-2, SOCS-3 and CIS mRNA and increased protein levels of SOCS-4 and SOCS-7 in AD brains. In summary, there was no evidence of a deficit of these key inflammatory regulating proteins in aged or AD brains.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Gene Expression/physiology , Microglia/metabolism , Signal Transduction/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Aged , Aged, 80 and over , Aging/pathology , Amyloid beta-Peptides/pharmacology , Brain/metabolism , Cognitive Dysfunction/pathology , Cytokines/genetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Female , Humans , Longitudinal Studies , Male , Microglia/drug effects , Organ Culture Techniques , Plaque, Amyloid/pathology , Suppressor of Cytokine Signaling Proteins/metabolism
3.
Neuroscience ; 302: 138-50, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-25281879

ABSTRACT

Microglia play major roles in initiation, coordination and execution of innate immunity in the brain. In the adult brain, these include maintenance of homeostasis, neuron and tissue repair, and eliminating infectious agents, apoptotic cells, and misfolded proteins. Some of these activities are accompanied by inflammatory reactions; and others are performed with no inflammatory effects. Under normal conditions, triggering receptor expressed on myeloid cells 2 (TREM2) belongs to the second category. It pairs with the adaptor protein DNAX-activating protein of 12kDa (DAP12) to induce phagocytosis of apoptotic neurons without inflammatory responses, and to regulate Toll-like receptor-mediated inflammatory responses, and microglial activation. Although ligands for TREM2 are largely unknown, the mitochondrial heat shock protein 60, expressed on cell surface of apoptotic neurons, is a specific ligand that activates TREM2-mediated phagocytosis by microglia. TREM2 also phagocytoses amyloid beta peptide in cultured cells. Several TREM2 mutations have been identified recently that increase the risk of Alzheimer's disease, Frontotemporal dementia, Parkinson's disease, and amyotrophic lateral sclerosis. Some of these mutations cause impaired proteolysis of full-length TREM2 at the plasma membrane to different degrees. The defects in the intramembrane cleavage result in dysfunction of phagocytosis signaling. The association of TREM2 mutations with neurodegenerative disease also calls for the understanding of the biology and pathological role of non-mutated TREM2 on human brains and microglia. This review provides a summary of current literature in TREM2 and DAP12 from several aspects, and proposes a theory that loss of TREM2 functions might contribute to the immunopathogenic role of microglia in Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Membrane Glycoproteins/genetics , Mutation/genetics , Receptors, Immunologic/genetics , Gene Expression Regulation/physiology , Humans , Microglia/metabolism , Microglia/pathology
4.
Curr Neuropharmacol ; 9(4): 693-705, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22654727

ABSTRACT

There is an urgent need for new ways to treat Alzheimer's disease (AD), the most common cause of dementia in the elderly. Current therapies are modestly effective at treating the symptoms, and do not significantly alter the course of the disease. Over the years, a range of epidemiological and experimental studies have demonstrated interactions between diabetes mellitus and AD. As both diseases are leading causes of morbidity and mortality in the elderly and are frequent co-morbid conditions, it has raised the possibility that treating diabetes might be effective in slowing AD. This is currently being attempted with drugs such as the insulin sensitizer rosiglitazone. These two diseases share many clinical and biochemical features, such as elevated oxidative stress, vascular dysfunction, amyloidogenesis and impaired glucose metabolism suggesting common pathogenic mechanisms. The main thrust of this review will be to explore the evidence from a pathological point of view to determine whether diabetes can cause or exacerbate AD. This was supported by a number of animal models of AD that have been shown to have enhanced pathology when diabetic conditions were induced. The one drawback in linking diabetes and insulin to AD has been the postmortem studies of diabetic brains demonstrating that AD pathology was not increased; in fact decreased pathology has often been reported. In addition, diabetes induces its own distinct features of neuropathology different from AD. There are common pathological features to be considered including vascular abnormalities, a major feature arising from diabetes; there is increasing evidence that vascular abnormalities can contribute to AD. The most important common mechanism between insulin-resistant (type II) diabetes and AD could be impaired insulin signaling; a form of toxic amyloid can damage neuronal insulin receptors and affect insulin signaling and cell survival. It has even been suggested that AD could be considered as "type 3 diabetes" since insulin can be produced in brain. Another common feature of diabetes and AD are increased advanced glycation endproduct-modified proteins are found in diabetes and in the AD brain; the receptor for advanced glycation endproducts plays a prominent role in both diseases. In addition, a major role for insulin degrading enzyme in the degradation of Aß peptide has been identified. Although clinical trials of certain types of diabetic medications for treatment of AD have been conducted, further understanding the common pathological processes of diabetes and AD are needed to determine whether these diseases share common therapeutic targets.

5.
Curr Drug Targets CNS Neurol Disord ; 4(3): 249-66, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15975028

ABSTRACT

Receptor for advanced glycation endproducts (RAGE), a member of the immunoglobulin superfamily, is a multi-ligand, cell surface receptor expressed by neurons, microglia, astrocytes, cerebral endothelial cells, pericytes, and smooth muscle cells. At least three major types of the RAGE isoforms (full length, C-truncated, and N-truncated) are present in human brains as a result of alternative splicing. Differential expression of each isoform may play a regulatory role in the physiological and pathophysiological functions of RAGE. Analysis of RAGE expression in non-demented and Alzheimer's disease (AD) brains indicated that increases in RAGE protein and percentage of RAGE-expressing microglia paralleled the severity of disease. Ligands for RAGE in AD include amyloid beta peptide (Abeta), S100/calgranulins, advanced glycation endproduct-modified proteins, and amphoterin. Collective evidence from in vitro and in vivo studies supports that RAGE plays multiple roles in the pathogenesis of AD. The major features of RAGE activation in contributing to AD result from its interaction with Abeta, from the positive feedback mechanisms driven by excess amounts of Abeta, and combined with sustained elevated RAGE expression. The adverse consequences of RAGE interaction with Abeta include perturbation of neuronal properties and functions, amplification of glial inflammatory responses, elevation of oxidative stress and amyloidosis, increased Abeta influx at the blood brain barrier and vascular dysfunction, and induction of autoantibodies. In this article, we will review recent advances of RAGE and RAGE activation based on findings from cell cultures, animal models, and human brains. The potential for targeting RAGE mechanisms as therapeutic strategies for AD will be discussed.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Glycation End Products, Advanced/metabolism , Microglia/metabolism , Receptors, Immunologic/metabolism , Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Animals , Blood Vessels/cytology , Blood Vessels/immunology , Blood Vessels/metabolism , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/immunology , Glycation End Products, Advanced/immunology , Humans , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microglia/immunology , Pericytes/immunology , Pericytes/metabolism , Protein Isoforms/metabolism , Receptor for Advanced Glycation End Products
6.
J Neurosci Res ; 81(3): 412-25, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15957156

ABSTRACT

Inflammation-mediated mechanisms for human neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) have evolved from being on the fringe of medical hypotheses to mainstream thinking. Pioneering immunopathology studies with human brain tissues identified microglia associated with neuropathologic hallmarks of these diseases. As activated macrophages were known to produce many potential toxic products, this gave rise to the hypothesis that activated microglia (brain resident macrophages) could be contributing to the degeneration of key target neurons in these diseases, as well as potential vascular dysfunction. Studies with microglia derived from different sources, including human brains, have confirmed that activated microglia can mediate neuronal cell death. Based on these theories, a number of human clinical trials with antiinflammatory agents have been carried out on AD patients. Results to date have indicated a lack of effectiveness at slowing disease progression and have begun to cast doubt on the significance of inflammation in AD. It has been shown recently that activating microglia through immunization of amyloid plaque-developing mice with amyloid beta peptide (Abeta) has promise as a therapeutic strategy and despite some setbacks, has potential as a treatment for AD patients. This article will consider experimental data with microglia to determine whether the additional targets need to be investigated. The use of human microglia cultures, in particular those derived from elderly diseased human brains, offers an experimental system that can closely model the cell type activated in human neurodegenerative diseases. Experimental data produced by our laboratory and others is reviewed to determine the contribution of this unique experimental model to understanding disease mechanisms and possibly discovering new therapeutic targets.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Microglia/pathology , Neurodegenerative Diseases/pathology , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Cells, Cultured , Endopeptidases/metabolism , Gene Expression/physiology , Humans , Inflammation/complications , Inflammation/pathology , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Species Specificity
8.
Neurochem Int ; 39(5-6): 333-40, 2001.
Article in English | MEDLINE | ID: mdl-11578768

ABSTRACT

Microglia are widely held to play important pathophysiologic roles in Alzheimer's disease (AD). On exposure to amyloid beta peptide (A beta) they exhibit chemotactic, phagocytic, phenotypic and secretory responses consistent with scavenger cell activity in a localized inflammatory setting. Because AD microglial chemotaxis, phagocytosis, and secretory activity have common, tightly linked soluble intermediaries (e.g., cytokines, chemokines), cell surface intermediaries (e.g., receptors, opsonins), and stimuli (e.g., highly inert A beta deposits and exposed neurofibrilly tangles), the mechanisms for microglial clearance of A beta are necessarily coupled to localized inflammatory mechanisms that can be cytotoxic to nearby tissue. This presents a critical dilemma for strategies to remove A beta by enhancing micoglial activation--a dilemma that warrants substantial further investigation.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Chemotaxis , Microglia/physiology , Phagocytosis , Humans
9.
Exp Neurol ; 171(1): 29-45, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11520119

ABSTRACT

Receptor-mediated interactions with amyloid beta-peptide (Abeta) could be important in the evolution of the inflammatory processes and cellular dysfunction that are prominent in Alzheimer's disease (AD) pathology. One candidate receptor is the receptor for advanced glycation endproducts (RAGE), which can bind Abeta and transduce signals leading to cellular activation. Data are presented showing a potential mechanism for Abeta activation of microglia that could be mediated by RAGE and macrophage colony-stimulating factor (M-CSF). Using brain tissue from AD and nondemented (ND) individuals, RAGE expression was shown to be present on microglia and neurons of the hippocampus, entorhinal cortex, and superior frontal gyrus. The presence of increased numbers of RAGE-immunoreactive microglia in AD led us to further analyze RAGE-related properties of these cells cultured from AD and ND brains. Direct addition of Abeta(1-42) to the microglia increased their expression of M-CSF. This effect was significantly greater in microglia derived from AD brains compared to those from ND brains. Increased M-CSF secretion was also demonstrated using a cell culture model of plaques whereby microglia were cultured in wells containing focal deposits of immobilized Abeta(1-42). In each case, the Abeta stimulation of M-CSF secretion was significantly blocked by treatment of cultures with anti-RAGE F(ab')2. Treatment of microglia with anti-RAGE F(ab')2 also inhibited the chemotactic response of microglia toward Abeta(1-42). Finally, incubation of microglia with M-CSF and Abeta increased expression of RAGE mRNA. These microglia also expressed M-CSF receptor mRNA. These data suggest a positive feedback loop in which Abeta-RAGE-mediated microglial activation enhances expression of M-CSF and RAGE, possibly initiating an ascending spiral of cellular activation.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Receptors, Immunologic/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Cells, Cultured , Chemotaxis/drug effects , Dose-Response Relationship, Drug , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Feedback/drug effects , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunoglobulin Fab Fragments/pharmacology , Immunohistochemistry , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Middle Aged , Neurons/metabolism , Neurons/pathology , Peptide Fragments/pharmacology , RNA, Messenger/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Signal Transduction
10.
Glia ; 35(1): 72-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11424194

ABSTRACT

We have previously developed and characterized isolated microglia and astrocyte cultures from rapid (<4 h) brain autopsies of Alzheimer's disease (AD) and nondemented elderly control (ND) patients. In the present study, we evaluate the inflammatory repertoire of AD and ND microglia cultured from white matter (corpus callosum) and gray matter (superior frontal gyrus) with respect to three major proinflammatory cytokines, three chemokines, a classical pathway complement component, a scavenger cell growth factor, and a reactive nitrogen intermediate. Significant, dose-dependent increases in the production of pro-interleukin-1beta (pro-IL-1beta), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-alpha), monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory peptide-1alpha (MIP-1alpha), IL-8, and macrophage colony-stimulating factor (M-CSF) were observed after exposure to pre-aggregated amyloid beta peptide (1-42) (Abeta1-42). Across constitutive and Abeta-stimulated conditions, secretion of complement component C1q, a reactive nitrogen intermediate, and M-CSF was significantly higher in AD compared with ND microglia. Taken together with previous in situ hybridization findings, these results demonstrate unequivocally that elderly human microglia provide a brain endogenous source for a wide range of inflammatory mediators.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , Brain/immunology , Encephalitis/immunology , Microglia/immunology , Aged , Aged, 80 and over , Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/pharmacology , Biomarkers/analysis , Brain/metabolism , Brain/physiopathology , Cells, Cultured/drug effects , Cells, Cultured/immunology , Cells, Cultured/metabolism , Chemokines/biosynthesis , Complement C1q/biosynthesis , Complement C1q/drug effects , Corpus Callosum/immunology , Corpus Callosum/metabolism , Corpus Callosum/physiopathology , Cytokines/biosynthesis , Cytokines/drug effects , Encephalitis/metabolism , Encephalitis/physiopathology , Female , Frontal Lobe/immunology , Frontal Lobe/metabolism , Frontal Lobe/physiopathology , Humans , Male , Microglia/drug effects , Microglia/metabolism , Nitrites/metabolism , Peptide Fragments/pharmacology
11.
Neurobiol Aging ; 22(6): 957-66, 2001.
Article in English | MEDLINE | ID: mdl-11755004

ABSTRACT

Activation of microglia is a central part of the chronic inflammatory processes in Alzheimer's disease (AD). In the brains of AD patients, activated microglia are associated with amyloid beta (Abeta) peptide plaques. A number of previous studies have shown that aggregated synthetic Abeta peptide activates cultured microglia to produce a range inflammatory products. The full extent of the inflammatory response still remains to be determined. In this study, gene array technology was employed to investigate in a more extensive manner the consequences of microglial activation by Abeta peptide. RNA was prepared from pooled samples of cortical human microglia isolated from post-mortem cases and incubated with a low dose (2.5 microM) of Abeta1-42 (or peptide solvent) for 24 h. This material was used to prepare cDNA probes, which were used to detect the differential pattern of expressed genes on a 1,176 Clontech membrane gene array. Results obtained showed that 104 genes were either upregulated or downregulated by 1.67 fold or greater. The most highly induced genes belonged to the chemokine family with interleukin-8 (IL-8) expression being increased by 11.7 fold. Interestingly, many of the highly induced genes had been identified as being responsive to activation by the transcription factor NF-kappaB. A number of genes were downregulated. Thymosin beta, prothymosin alpha and parathymosin, all belonging to the same gene family, were downregulated. To validate these semi-quantitative results, the expression of intercellular adhesion molecule-1 (ICAM-1) and rhoB were measured by RT-PCR in samples of cDNA derived from Abeta and control stimulated human cortical microglia. These results confirm the usefulness of the gene array approach for studying Abeta-mediated inflammatory processes.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/pharmacology , Gene Expression/drug effects , Microglia/metabolism , Alzheimer Disease/metabolism , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cells, Cultured , Chemokines/biosynthesis , Chemokines/genetics , Down-Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Microglia/drug effects , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical , rhoA GTP-Binding Protein/metabolism
12.
Neurobiol Aging ; 22(6): 945-56, 2001.
Article in English | MEDLINE | ID: mdl-11755003

ABSTRACT

Alzheimer's disease (AD) is a uniquely human disorder. Despite intense research, the lack of availability of model systems has hindered AD studies though in recent years transgenic mouse models have been produced, which develop AD-like amyloid beta peptide (Abeta) plaques. For the study of inflammatory changes in AD brains, these transgenic mice may have limitations due to differences in the innate immune system of humans and rodents. Many studies of inflammatory processes in AD have focused on the role of activated microglia. Over the last 8 years, our research has focused on the properties of human microglia cultured from brain tissues of AD and non-demented (ND) individuals. As these are the cells observed to be activated in AD tissues, they represent a useful system for modeling the inflammatory components of AD. In this review, we summarize data by our group and others on the use of microglia for AD-related inflammatory research, with emphasis on results using human postmortem brain microglia. A range of products have been shown to be produced by human postmortem microglia, both constitutively and in response to treatment with Abeta, including proinflammatory cytokines such as interleukin (IL)-1beta, IL-6, tumor necrosis factor (TNF) alpha, and macrophage colony stimulating factor (M-CSF), along with complement proteins, especially C1q, superoxide radicals and neurotoxic factors. In our studies, we have demonstrated that there was a significant difference between AD and ND microglia in terms of their secretion of M-CSF and C1q. We also discuss the role of putative Abeta microglial receptors, particular recent data showing a role for the receptor for advanced glycation endproducts (RAGE) in mediating the responses of human microglia to Abeta. Finally, our studies on the use of an Abeta spot paradigm to model microglia interactions with plaques demonstrated that many of the features of AD inflammation can be modeled with postmortem brain derived microglia.


Subject(s)
Alzheimer Disease/pathology , Enzyme Activation/physiology , Microglia/physiology , Animals , Brain/cytology , Cells, Cultured , Humans , Models, Neurological , Nerve Tissue Proteins/metabolism , Plaque, Amyloid/pathology
13.
J Neurosci Methods ; 103(1): 3-10, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11074091

ABSTRACT

The use of Di-I in tract-tracing is briefly reviewed and a novel delayed-fixation approach to neural tract-tracing in the postmortem human adult brain is reported. Using the new approach, fast Di-I, a highly lipophilic fluorescent dye was injected into a particular region or nucleus and labelled tracts were followed for distances of some 20-40 mm. The procedure required approximately 36 h, yielding dye penetration rates of 1.0 mm/h or more. This contrasts with previous Di-I, silver impregnation, and horseradish peroxidase protocols, where the tracer penetration rate is typically 0.003 mm/h or less, and the distance traversed amounts to only a few mm even after months of incubation. The new method hinges on the simple consideration that aldehyde fixation, which is normally employed prior to administration of the marker, crosslinks membrane proteins and impedes dye diffusion. The short postmortem samples used in our protocol permit delaying fixation until after the dye has had time to penetrate, dramatically increasing the length and scope of neural circuits that can be traced. Using these methods, for example, we have confirmed the presence of an ipsilateral olivocerebellar climbing fiber projection in the human.


Subject(s)
Axons/ultrastructure , Brain/cytology , Carbocyanines , Fluorescent Dyes , Nerve Net/cytology , Neural Pathways/cytology , Animals , Axons/physiology , Brain/physiology , Diffusion/drug effects , Humans , Injections/methods , Nerve Net/physiology , Neural Pathways/physiology , Tissue Fixation/methods , Tissue Fixation/standards
14.
J Neurochem ; 75(4): 1447-54, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10987824

ABSTRACT

In recent years, inflammatory mechanisms have been increasingly appreciated as important steps in the pathology of Alzheimer's disease (AD). There are two pathological defects in AD: chronic inflammation and impaired clearance of amyloid beta-peptide (Abeta). In the periphery, estrogen both increases macrophage phagocytosis and has antiinflammatory effects. If estrogen had a similar effect in the CNS, it could reverse inflammatory defects in AD. Although microglia are a key component of the immune system and help clear Abeta deposits in the AD brain, little is known about the effects of estrogen on CNS microglia. Therefore, we sought to determine the relationship between estrogen treatment and internalization of Abeta by microglia by quantifying the internalization of aggregated Abeta by human cortical microglia. Abeta uptake was found to be dose- and time-dependent in cultured microglia. Increased Abeta uptake was observed at 1.5 and 24 h after addition of aggregated Abeta (50, 100, or 1,000 nM: Abeta), and this uptake was enhanced by pretreatment with estrogen. The expression of estrogen receptor (ER) beta (ER-beta) was also up-regulated by estrogen treatment. Cells cotreated with ICI 182,780, an ER antagonist, showed significantly reduced internalization of Abeta in cultured microglia. These results indicate that microglia express an ER-beta but that the effect of estrogen on enhancing clearance of Abeta may be related to the receptor-independent action of estrogen or to nonclassical ER effects of estrogen. Thus, stimulation of the ER might contribute to the therapeutic action of estrogen in the treatment of AD.


Subject(s)
Amyloid beta-Peptides/pharmacokinetics , Estradiol/analogs & derivatives , Estradiol/pharmacology , Frontal Lobe/drug effects , Microglia/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Estrogen Antagonists/pharmacology , Estrogens/metabolism , Fluorescent Dyes , Frontal Lobe/cytology , Frontal Lobe/metabolism , Fulvestrant , Humans , Immunohistochemistry , Microglia/cytology , Microglia/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism
15.
Am J Pathol ; 155(3): 853-62, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10487842

ABSTRACT

We have characterized amyloid beta peptide (Abeta) concentration, Abeta deposition, paired helical filament formation, cerebrovascular amyloid angiopathy, apolipoprotein E (ApoE) allotype, and synaptophysin concentration in entorhinal cortex and superior frontal gyrus of normal elderly control (ND) patients, Alzheimer's disease (AD) patients, and high pathology control (HPC) patients who meet pathological criteria for AD but show no synapse loss or overt antemortem symptoms of dementia. The measures of Abeta deposition, Abeta-immunoreactive plaques with and without cores, thioflavin histofluorescent plaques, and concentrations of insoluble Abeta, failed to distinguish HPC from AD patients and were poor correlates of synaptic change. By contrast, concentrations of soluble Abeta clearly distinguished HPC from AD patients and were a strong inverse correlate of synapse loss. Further investigation revealed that Abeta40, whether in soluble or insoluble form, was a particularly useful measure for classifying ND, HPC, and AD patients compared with Abeta42. Abeta40 is known to be elevated in cerebrovascular amyloid deposits, and Abeta40 (but not Abeta42) levels, cerebrovascular amyloid angiopathy, and ApoE4 allele frequency were all highly correlated with each other. Although paired helical filaments in the form of neurofibrillary tangles or a penumbra of neurites surrounding amyloid cores also distinguished HPC from AD patients, they were less robust predictors of synapse change compared with soluble Abeta, particularly soluble Abeta40. Previous experiments attempting to relate Abeta deposition to the neurodegeneration that underlies AD dementia may have failed because they assayed the classical, visible forms of the molecule, insoluble neuropil plaques, rather than the soluble, unseen forms of the molecule.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Synapses/pathology , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Apolipoproteins E/genetics , Brain/metabolism , Brain/pathology , Cerebral Amyloid Angiopathy/pathology , Diagnosis, Differential , Female , Gene Frequency , Humans , Immunohistochemistry , Male , Neurofibrillary Tangles/pathology , Peptide Fragments/metabolism , Plaque, Amyloid/pathology , Predictive Value of Tests
16.
Neurobiol Aging ; 18(4): 415-21, 1997.
Article in English | MEDLINE | ID: mdl-9330973

ABSTRACT

The membrane attack complex, C5b-9, is of considerable importance in many inflammatory reactions. It is the terminal, cytolytic component of both classical and alternative pathway activation, and its presence presupposes other potentially destructive complement constituents, including anaphylotoxins and opsonins. We have characterized C5b-9 and its C9 constituent in the Alzheimer's disease (AD) and nondemented elderly (ND) brain using immunohistochemistry at the light and electron microscopic levels, Western blot analysis, and the reverse transcriptase polymerase chain reaction. We have also conducted in vitro ELISA assays of amyloid beta-peptide-stimulated SC5b-9 production. C5b-9 is abundantly present in Alzheimer's disease cortex, associated with neurofibrillary tangle containing neurons, dystrophic neurites within neuritic plaques, and neuropil threads, but is weakly detected, if at all, in nondemented elderly cortex under the same conditions. Staining of Alzheimer's disease sections is abolished both by deletion of primary antibody or preabsorption with purified SC5b-9.


Subject(s)
Alzheimer Disease/metabolism , Brain Chemistry , Complement Membrane Attack Complex/analysis , Base Sequence , Blotting, Western , Brain/metabolism , Complement Membrane Attack Complex/chemistry , Frontal Lobe/chemistry , Frontal Lobe/metabolism , Humans , Immunohistochemistry , Molecular Sequence Data , Neurofibrillary Tangles/metabolism , Polymerase Chain Reaction , Temporal Lobe/chemistry , Temporal Lobe/metabolism
17.
J Neuropathol Exp Neurol ; 55(10): 1083-8, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8858005

ABSTRACT

We evaluated entorhinal cortex and superior frontal gyrus for hallmarks of Alzheimer's disease (AD) pathology, including inflammation, in three patient sets: AD patients, nondemented elderly patients with few or no neurofibrillary tangles (NFTs) and amyloid beta peptide (A beta) deposits, i.e. normal controls (NC), and nondemented elderly patients with profuse entorhinal cortex NFTs and neocortical A beta deposits, i.e. high pathology controls (HPC). Membrane attack complex (C5b-9) immunoreactivity and immune activation of microglia (MHCII expression) were used as general markers for inflammation. Compared to NC patients, AD patients exhibited significant cortical synapse loss, A beta deposition, NFT formation, and inflammation. HPC patients also had significantly elevated A beta deposition and NFT formation, but there was no evidence of synapse loss and little or no evidence of inflammation. Across patients and brain regions the measures of inflammation each accounted for significant percentages of the variance in synaptophysin immunoreactivity and each was more highly correlated with synapse estimates than NFT formation or A beta deposition.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/analysis , Neuritis/pathology , Neurofibrillary Tangles/chemistry , Aged , Aged, 80 and over , Alzheimer Disease/immunology , Biomarkers , Blotting, Western , Entorhinal Cortex/chemistry , Entorhinal Cortex/immunology , Entorhinal Cortex/pathology , Female , Frontal Lobe/chemistry , Frontal Lobe/immunology , Frontal Lobe/pathology , Histocytochemistry , Humans , Immunohistochemistry , Male , Microglia/chemistry , Nerve Degeneration/physiology , Neurofibrillary Tangles/immunology
18.
Neurobiol Aging ; 17(5): 681-6, 1996.
Article in English | MEDLINE | ID: mdl-8892340

ABSTRACT

Appreciation of the role that inflammatory mediators play in Alzheimer's disease (AD) pathogenesis continues to be hampered by two related misconceptions. The first is that to be pathogenically significant a neurodegenerative mechanism must be primary. The second is that inflammation merely occurs to clear the detritis of already existent pathology. The present review addresses these issues by showing that 1) inflammatory molecules and mechanisms are uniquely present or significantly elevated in the AD brain, 2) inflammation may be a necessary component of AD pathogenesis, 3) inflammation may be sufficient to cause AD neurodegeneration, and 4) retrospective and direct clinical trials suggest a therapeutic benefit of conventional antiinflammatory medications in slowing the progress or even delaying the onset of AD.


Subject(s)
Alzheimer Disease/pathology , Inflammation/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Chemistry/drug effects , Humans , Inflammation/metabolism , Inflammation Mediators/physiology
19.
Neurobiol Aging ; 17(3): 421-9, 1996.
Article in English | MEDLINE | ID: mdl-8725904

ABSTRACT

We have developed isolated and mixed cultures of microglia, astrocytes, and oligodendrocytes from rapid (mean of 2 h 55 min) autopsies of nondemented elderly patients and patients with Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Cultures were derived from both the corpus callosum (CC) and superior frontal gyrus (SFG). Cultured microglia phagocytosed latex beads, were reactive for Dil-acetylated low density lipoprotein, were immunoreactive for CD68 and major histocompatibility complex II markers, and were not immunoreactive for fibroblast, astrocyte, or oligodendrocyte markers. Cultured astrocytes included fibrous and protoplasmic types, were immunoreactive for GFAP, and were not immunoreactive for fibroblast, microglia, or oligodendrocyte markers. Cultured oligodendrocytes were poorly adherent, were slow to develop, were immunoreactive for galactocerebroside, and were not immunoreactive for fibroblast, microglia, or astrocyte markers. Because they are readily manipulated under controlled experimental conditions, and because they permit immediate access to individual cells and sets of cells from patients who have actually suffered the disease, these cultures may provide an important new tool for unravelling the etiology and pathogenesis of human CNS disorders.


Subject(s)
Alzheimer Disease/pathology , Astrocytes/pathology , Microglia/pathology , Neuroglia/pathology , Aged , Aged, 80 and over , Cells, Cultured/pathology , Female , Humans , Immunohistochemistry , Male
20.
Brain Res Mol Brain Res ; 18(4): 329-34, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8326828

ABSTRACT

Concentrations of C1q, the first subcomponent of the classical complement pathway, were assayed by Western blot analysis of sera and brain homogenates from Alzheimer's disease (AD) and nondemented (ND) control patients. Immunoreactive serum C1q concentrations did not differ in the two groups, whereas AD superior frontal gyrus exhibited nearly 4-fold more immunoreactive C1q than ND superior frontal gyrus. Cerebellar C1q concentrations were significantly lower than those in superior frontal gyrus, and ND cerebellar C1q was lowest of all. Parallel immunohistochemical experiments showed a linkage between the extent of beta-amyloid immunoreactivity or AD pathology in a structure and the extent of C1q immunoreactivity. These data support and extend the hypothesis that complement mediated processes are related to beta-amyloid deposition and may be involved in the pathogenesis of AD.


Subject(s)
Alzheimer Disease/immunology , Cerebellum/immunology , Cerebral Cortex/immunology , Complement C1q/metabolism , Complement Factor B/metabolism , Adult , Aged , Aged, 80 and over , Humans , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...