Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Brain Pathol ; 29(5): 606-621, 2019 09.
Article in English | MEDLINE | ID: mdl-30661261

ABSTRACT

Among the changes that typify Alzheimer's disease (AD) are neuroinflammation and microglial activation, amyloid deposition perhaps resulting from compromised microglial function and iron accumulation. Data from Genome Wide Association Studies (GWAS) identified a number of gene variants that endow a significant risk of developing AD and several of these encode proteins expressed in microglia and proteins that are implicated in the immune response. This suggests that neuroinflammation and the accompanying microglial activation are likely to contribute to the pathogenesis of the disease. The trigger(s) leading to these changes remain to be identified. In this study, we set out to examine the link between the inflammatory, metabolic and iron-retentive signature of microglia in vitro and in transgenic mice that overexpress the amyloid precursor protein (APP) and presenilin 1 (PS1; APP/PS1 mice), a commonly used animal model of AD. Stimulation of cultured microglia with interferon (IFN)γ and amyloid-ß (Aß) induced an inflammatory phenotype and switched the metabolic profile and iron handling of microglia so that the cells became glycolytic and iron retentive, and the phagocytic and chemotactic function of the cells was reduced. Analysis of APP/PS1 mice by magnetic resonance imaging (MRI) revealed genotype-related hypointense areas in the hippocampus consistent with iron deposition, and immunohistochemical analysis indicated that the iron accumulated in microglia, particularly in microglia that decorated Aß deposits. Isolated microglia prepared from APP/PS1 mice were characterized by a switch to a glycolytic and iron-retentive phenotype and phagocytosis of Aß was reduced in these cells. This evidence suggests that the switch to glycolysis in microglia may kick-start a cascade of events that ultimately leads to microglial dysfunction and Aß accumulation.


Subject(s)
Alzheimer Disease/metabolism , Iron/metabolism , Microglia/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/pathology , Cytokines/metabolism , Disease Models, Animal , Female , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism
2.
Neurosci Res ; 148: 54-60, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30641113

ABSTRACT

The search for a blood-based biomarker that identifies Alzheimer's disease (AD) and can replace current invasive and expensive diagnostic tests, continues. The most extensively-examined peripheral marker is ß-amyloid (Aß) but the results are inconsistent across studies and do not reflect the changes that take place in the brain. Several studies have assessed possible proteomic signatures but with inconsistent findings, although increases in circulating inflammatory molecules are generally observed. Here, rather than focus on identifying changes in the circulation, we evaluated the effect of plasma from patients with mild cognitive impairment (MCI) and AD on the human monocyte-like cell line, THP-1 cells, and plasma from an AD mouse model on a mouse monocyte-macrophage cell line, J774.2 cells. Plasma from AD patients and the AD mouse model increased inflammatory molecules in the cells and these changes were accompanied by an increase in glycolysis. Interestingly, plasma from MCI patients exerted no significant effect on THP-1 cells. The possibility therefore exists that evaluating the effect of plasma on IL-8 and TNFα mRNA in THP-1 cells combined with analysis of glycolysis in these cells, may be the basis of an indicator that discriminates between AD and MCI and normal controls, but is unlikely to be useful in identifying early pathological changes.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Monocytes/metabolism , Aged , Amyloid beta-Peptides/metabolism , Animals , Chemokine CXCL1/metabolism , Cytokines/metabolism , Female , Glycolysis , Humans , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Nitric Oxide Synthase Type II/metabolism , THP-1 Cells
3.
Nat Nanotechnol ; 13(5): 427-433, 2018 05.
Article in English | MEDLINE | ID: mdl-29610530

ABSTRACT

The potential for maternal nanoparticle (NP) exposures to cause developmental toxicity in the fetus without the direct passage of NPs has previously been shown, but the mechanism remained elusive. We now demonstrate that exposure of cobalt and chromium NPs to BeWo cell barriers, an in vitro model of the human placenta, triggers impairment of the autophagic flux and release of interleukin-6. This contributes to the altered differentiation of human neural progenitor cells and DNA damage in the derived neurons and astrocytes. Crucially, neuronal DNA damage is mediated by astrocytes. Inhibiting the autophagic degradation in the BeWo barrier by overexpression of the dominant-negative human ATG4BC74A significantly reduces the levels of DNA damage in astrocytes. In vivo, indirect NP toxicity in mice results in neurodevelopmental abnormalities with reactive astrogliosis and increased DNA damage in the fetal hippocampus. Our results demonstrate the potential importance of autophagy to elicit NP toxicity and the risk of indirect developmental neurotoxicity after maternal NP exposure.


Subject(s)
Astrocytes/metabolism , Models, Biological , Nanoparticles/toxicity , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Placenta/pathology , Pregnancy Complications/metabolism , Animals , Astrocytes/pathology , Cell Line , Female , Humans , Male , Mice , Neurodevelopmental Disorders/chemically induced , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neurons/pathology , Neurotoxicity Syndromes/pathology , Pregnancy , Pregnancy Complications/chemically induced , Pregnancy Complications/pathology
4.
Prog Neuropsychopharmacol Biol Psychiatry ; 79(Pt A): 15-18, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28189704

ABSTRACT

Peripheral macrophages have limited capacity to gain access to the brain parenchyma under normal physiological conditions. However, accumulating evidence indicates that significant trafficking to the central nervous systems occurs in response to injury or infection and is also apparent under chronic neuroinflammatory conditions. The role of infiltrating macrophages in neuronal function is unclear and confounded by the similarity in morphology and phenotype adopted by both activated macrophages and microglia. Furthermore, the ability of macrophages/microglia to adopt both pro- and anti-inflammatory activation states, along with the fact that these cells display heterogenous expression of molecules associated with both states, has made it difficult to discover their impact upon neuronal injury and cognitive processes. The ability of macrophages to exert a neuroprotective role is influenced by the microenvironment they encounter upon tissue invasion. Upon encountering an inflammatory microenvironment, macrophage polarisation is driven towards a pro-inflammatory (M1) phenotype, a state associated with reduced capacity for restorative processes such as the removal of debris, and enhanced production of pro-inflammatory mediators such as TNFα, IL-1ß and NADPH oxidase. Prolonged production of these inflammatory mediators has been shown to affect neuronal function and health. Thus, macrophage polarisation may be dictated by the inflammatory queues these cells are exposed to upon migration and their subsequent impact on neuronal function may be determined by their ability to resolve the underlying inflammation.


Subject(s)
Encephalitis/complications , Encephalitis/pathology , Macrophages/physiology , Monocytes/physiology , Affect/physiology , Animals , Cognition/physiology , Cytokines/metabolism , Humans , Learning/physiology
5.
Mol Neurobiol ; 54(7): 5730-5739, 2017 09.
Article in English | MEDLINE | ID: mdl-27830533

ABSTRACT

One factor that impacts on microglial activation is the interaction between the ubiquitously expressed CD200 and CD200R, which is expressed only on microglia in the brain. Decreased signalling through CD200R, when CD200 expression is reduced, results in microglial activation and may, at least in part, explain the increased cell activity that is observed with age, in models of Alzheimer's and Parkinson's disease as well as in the human diseases. There is evidence of increased microglial activation in CD200-deficient mice, and isolated microglia prepared from these mice are more reactive to inflammatory stimuli like Toll-like receptor 2 and 4 agonists, and interferon-γ. Here, we examined the impact of CD200 deficiency on amyloid-ß (Aß)-induced changes in microglia and report, perhaps unexpectedly, that the effect of Aß was attenuated in microglia prepared from CD200-deficient mice. The evidence indicates that this is a consequence of increased phagocytosis, associated with increased lysosomal activity in CD200-deficient microglia. The data suggest that mTOR-related signalling is decreased in these cells and that inhibiting mTOR by rapamycin increases phagocytosis. Thus, while the findings to date have emphasized the anti-inflammatory effects of CD200-CD200R interaction, the present evidence indicates a previously unreported impact on lysosomal function.


Subject(s)
Antigens, CD/metabolism , Lysosomes/metabolism , Microglia/metabolism , Phagocytosis/physiology , Amyloid beta-Peptides/metabolism , Animals , Hippocampus/metabolism , Interferon-gamma/metabolism , Macrophage Activation/physiology , Male , Microglia/drug effects , Rats, Wistar , Toll-Like Receptor 2/metabolism
6.
Neurobiol Aging ; 36(10): 2716-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26227742

ABSTRACT

There is a wealth of evidence indicating that macrophages adopt distinct phenotypes when exposed to specific stimuli and, in the past few years, accumulating data suggest that microglia behave somewhat similarly. Therefore, microglia can adopt the so-called M1 or M2 phenotypes in response to interferon-γ (IFNγ) and interleukin-4, respectively. Although it has yet to be unequivocally proven in the context of microglia, acutely activated M1 cells are probably protective, although a persistent M1 state is likely to be damaging, whereas M2 cells may be reparative and restorative. In this case, particularly because the current evidence suggests the development of a predominantly M1 state with age and in neurodegenerative diseases, it is important to identify mechanisms by which polarization of microglia can be modulated. The present findings indicate that exposure of cultured microglia to IFNγ increased expressions of the archetypal markers of the M1 phenotype, tumour necrosis factor-α, and inducible nitric oxide synthase, and preexposure of cells to amyloid-ß (Aß) sensitized microglia to subsequent stimulation with IFNγ. Importantly, this synergy was also evident in microglia prepared from the brains of transgenic mice that overexpress amyloid precursor protein (APP) and presenilin 1 (PS1, APP/PS1 mice) and are exposed to a combination of increasing concentrations of endogenous Aß from 4 or 5 months of age and an age-related increase in IFNγ. Significantly, the JAK2 inhibitor, TG101209, attenuated the IFNγ-induced changes in cultured microglia and in isolated microglia prepared from APP/PS1 mice. These findings suggest that targeting JAK2 may be a potential strategy for reducing neuroinflammation in Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Inflammation Mediators/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/physiology , Microglia/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Alzheimer Disease/etiology , Amyloid beta-Peptides , Animals , Cells, Cultured , Interferon-gamma/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Nitric Oxide Synthase Type II , Presenilin-1 , Tumor Necrosis Factor-alpha
7.
Exp Cell Res ; 335(2): 258-68, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26022664

ABSTRACT

Macrophages can be polarised to adopt the M1 or M2 phenotype and functional outcomes of activation include altered secretion of immune molecules such as insulin-like growth factor (IGF)-1 as well as upregulation of cell surface molecules specifically associated with each state. Interleukin (IL)-4 mediates its effects through two receptors, the type I and II receptors and activation of these receptors results in phosphorylation of signal transducers and activators of transcription (STAT)6. JAK3 is activated as a consequence of ligation of the type I IL-4R, which participates in Akt activation. We set out to investigate the impact of perturbation of IGF-1 tone on IL-4- and interferon (IFN)γ-induced activation, the mechanisms by which this may occur and the contribution of type I IL-4R activation to adoption of the M2 state. The data presented here indicate that IL-4-induced activation of Akt is JAK3-dependent, enhanced by release of IGF-1 and necessary for full adoption of the M2 phenotype, since blocking IGF-1 activity blunts the ability of IL-4 to induce activation of Akt and to upregulate expression of some M2-associated molecules. In addition, differential control of the expression of mannose receptor (MRC1), arginase-1 (Arg-1), chitinase-3 like 3 (Chi3l3) and found in inflammatory zone 1 (FIZZ1) was observed. The IFNγ-induced decrease in IGF-1 was exacerbated by inhibition of phosphatidylinositol-3 (PI3) kinase, indicating that Akt may regulate its own activation via IGF-1. Overall, a deficit in IGF-1/Akt signalling is associated with decreased capacity to induce the M2 state and an increased responsiveness to IFNγ.


Subject(s)
Insulin-Like Growth Factor I/physiology , Macrophages/enzymology , Proto-Oncogene Proteins c-akt/physiology , Animals , Cells, Cultured , Interleukin-4/physiology , Janus Kinase 3/metabolism , Macrophage Activation , Macrophages/immunology , Mice , Signal Transduction , Transcriptome
8.
J Alzheimers Dis ; 44(3): 949-62, 2015.
Article in English | MEDLINE | ID: mdl-25374107

ABSTRACT

Macrophages are key cells in tissue defense in the periphery and, under certain circumstances, infiltrate the central nervous system, where they may play a similar role in the brain, perhaps supporting the function of microglia. Macrophages have been shown to adopt different activation states in response to various stimuli. Specifically, when exposed to inflammatory stimuli such as interferon (IFN)γ, the cells adopt the M1 phenotype, whereas when exposed to anti-inflammatory cytokines such as interleukin (IL)-4 or IL-13, the M2 phenotype is adopted. While M1 macrophages are associated with tissue defense and destruction of invading pathogens, M2 macrophages are involved in tissue repair and in terminating inflammation. It is well known that an inflammatory microenvironment exists in the brain of aged animals and also in the brain of mice that overexpress amyloid-ß protein precursor (AßPP) and presenilin 1 (PS1; AßPP/PS1 mice), a commonly-used model of Alzheimer's disease (AD). Recent studies have revealed that immune cells, including macrophages, infiltrate the brain in both circumstances raising the possibility that these cells adopt the M1 activation state and contribute to the already-existing neuroinflammation. We set out to examine the responses of bone marrow-derived macrophages prepared from wildtype and AßPP/PS1 mice and demonstrate that cells from AßPP/PS1 mice, even after several days in culture, respond more profoundly to IFNγ than those from wildtype mice. We suggest that this propensity to respond to M1-polarizing stimuli, together with the described changes in the brain of AßPP/PS1 mice, contribute to the development of chronic neuroinflammation.


Subject(s)
Alzheimer Disease/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Interferon-gamma/pharmacology , Macrophages/drug effects , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Claudin-5/metabolism , Cytokines/metabolism , Disease Models, Animal , Endothelium/drug effects , Female , Humans , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Presenilin-1/genetics , Receptors, Cell Surface/metabolism , Receptors, Immunologic , Zonula Occludens-1 Protein/metabolism
9.
Neurobiol Aging ; 35(6): 1442-52, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24439957

ABSTRACT

Aging adversely affects inflammatory processes in the brain, which has important implications in the context of disease progression. It has been proposed that microglia become dysfunctional with age and may lose their neuroprotective properties leading to chronic neurodegeneration. Here, we sought to characterize inflammatory changes in a mouse model of Alzheimer's disease and to delineate differences between normal aging and those associated with disease pathology. A proinflammatory profile, characterized by the upregulation of markers of classical activation, was evident in APPswe/PS1dE9 mice, associated with increased interferon-γ (IFNγ) concentration and dysregulation of mechanisms designed to limit the proinflammatory response. The data indicate that microglia are not less active with age but alter their phenotype; indeed, changes observed in the deactivation state appear to relate to aging rather than disease pathology. We hypothesize that disruption of the blood-brain barrier, in tandem with an enhanced chemokine profile, permits the infiltration of immune cells serving to reinforce classical activation of microglia through their enhanced responsiveness to IFNγ.


Subject(s)
Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Blood-Brain Barrier/metabolism , Microglia/pathology , Mutation , Presenilin-1/genetics , Alzheimer Disease/genetics , Animals , Blood-Brain Barrier/pathology , Disease Models, Animal , Female , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Permeability , Up-Regulation
10.
J Alzheimers Dis ; 37(1): 63-75, 2013.
Article in English | MEDLINE | ID: mdl-23780663

ABSTRACT

Whereas the classical histological hallmarks of Alzheimer's disease (AD) are deposition of amyloid-containing plaques and development of neurofibrillary tangles, there is also clear evidence of inflammatory changes accompanied by the presence of activated microglia and astrocytosis. However, at this time, it remains uncertain whether inflammatory changes contribute to pathogenesis of the disease or if they are secondary to deposition of amyloid-ß or other pathological changes. A greater understanding of the sequence of events would clearly improve development of strategies to delay progression of the disease. There is a realistic expectation that advances in imaging technology may provide the key to uncovering this sequence. In this study, we employed non-invasive imaging techniques to examine changes in tissue state in hippocampus and cortex of transgenic mice which overexpress amyloid-ß protein precursor and presenilin 1 and show that the observed increase in T1 relaxation time was associated with astrogliosis while the decrease in T2 relaxation time was associated with microglial activation. We explored the possibility that interferon-γ might trigger glial activation and demonstrate a genotype-related infiltration of macrophages and natural killer cells, which release interferon-γ. The evidence suggests that IFNγ triggers glial activation and expression of proinflammatory cytokines, and these changes, in turn, contribute to the decrease in long-term potentiation.

11.
J Neuroimmune Pharmacol ; 8(1): 301-11, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23238794

ABSTRACT

Astrocytes, the most numerous glial cell in the brain, have multiple functions and are key to maintenance of homeostasis in the central nervous system. Microglia are the resident immunocompetent cells in the brain and share several functions with macrophages, including their phagocytic ability. Indeed microglia are the resident phagocytes in the brain and express numerous cell surface proteins which act to enable receptor-mediated phagocytosis. However recent evidence suggests that astrocytes express some genes which permit phagocytosis of phosphatidylserine-decorated cells and this probably explains sporadic reports in the literature which suggest that astrocytes become phagocytic following brain trauma. Here we examined the potential of astrocytes to phagocytose fluorescently-labelled latex beads and amyloid-ß (Aß) and report that they competently engulf both in a manner that relies on actin polymerization since it was inhibited by cytochalasin D. The data indicate that incubation of cultured astrocytes or microglia with Aß increased phagocytosis and markers of activation of both cell types. Aß was found to markedly increase expression of the putative Aß-binding receptors CD36 and CD47 in astrocytes, while it decreased expression of the receptor for advanced glycation endproducts (RAGE). It is demonstrated that blocking these receptors using a neutralizing antibody attenuated Aß-induced phagocytosis of latex beads by astrocytes. Interestingly blocking these receptors also decreased uptake of beads even in the absence of Aß. Here we demonstrate that astrocytes are competent phagocytes and are capable of engulfing Aß.


Subject(s)
Amyloid beta-Peptides/toxicity , Astrocytes/drug effects , CD36 Antigens/physiology , CD47 Antigen/physiology , Phagocytosis/drug effects , Receptors, Immunologic/physiology , Animals , Animals, Newborn , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Cell Survival/drug effects , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Flow Cytometry , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , L-Lactate Dehydrogenase/metabolism , Microglia/drug effects , Microspheres , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products
12.
J Neuroinflammation ; 9: 126, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22697788

ABSTRACT

BACKGROUND: Compelling evidence has implicated neuroinflammation in the pathogenesis of a number of neurodegenerative conditions. Chronic activation of both astrocytes and microglia leads to excessive secretion of proinflammatory molecules such as TNF α, IL-6 and IL-1 ß with potentially deleterious consequences for neuronal viability. Many signaling pathways involving the mitogen-activated protein kinases (MAPKs), nuclear factor κ B (NF κ B) complex and the Janus kinases (JAKs)/signal transducers and activators of transcription (STAT)-1 have been implicated in the secretion of proinflammatory cytokines from glia. We sought to identify signaling kinases responsible for cytokine production and to delineate the complex interactions which govern time-related responses to lipopolysaccharide (LPS). METHODS: We examined the time-related changes in certain signaling events and the release of proinflammatory cytokines from LPS-stimulated co-cultures of astrocytes and microglia isolated from neonatal rats. RESULTS: TNF α was detected in the supernatant approximately 1 to 2 hours after LPS treatment while IL-1 ß and IL-6 were detected after 2 to 3 and 4 to 6 hours, respectively. Interestingly, activation of NF κ B signaling preceded release of all cytokines while phosphorylation of STAT1 was evident only after 2 hours, indicating that activation of JAK/STAT may be important in the up-regulation of IL-6 production. Additionally, incubation of glia with TNF α induced both phosphorylation of JAK2 and STAT1 and the interaction of JAK2 with the TNF α receptor (TNFR1). Co-treatment of glia with LPS and recombinant IL-6 protein attenuated the LPS-induced release of both TNF α and IL-1 ß while potentiating the effect of LPS on suppressor of cytokine signaling (SOCS)3 expression and IL-10 release. CONCLUSIONS: These data indicate that TNF α may regulate IL-6 production through activation of JAK/STAT signaling and that the subsequent production of IL-6 may impact on the release of TNF α, IL-1 ß and IL-10.


Subject(s)
Cytokine Receptor gp130/metabolism , Interleukin-1beta/biosynthesis , Janus Kinase 2/physiology , Lipopolysaccharides/physiology , Neuroglia/metabolism , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , Cytokine Receptor gp130/physiology , MAP Kinase Signaling System/physiology , Neuroglia/immunology , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Signal Transduction/physiology
13.
FEBS J ; 277(6): 1503-18, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20163459

ABSTRACT

The amyloid precursor family of proteins are of considerable interest, both because of their role in Alzheimer's disease pathogenesis and because of their normal physiological functions. In mammals, the amyloid precursor protein (APP) has two homologs, amyloid precursor-like protein (APLP) 1 and APLP2. All three proteins undergo ectodomain shedding and regulated intramembrane proteolysis, and important functions have been attributed to the full-length proteins, shed ectodomains, C-terminal fragments and intracellular domains (ICDs). One of the proteases that is known to cleave APP and that is essential for generation of the amyloid beta-protein is the beta-site APP-cleaving enzyme 1 (BACE1). Here, we investigated the effects of genetic manipulation of BACE1 on the processing of the APP family of proteins. BACE1 expression regulated the levels and species of full-length APLP1, APP and APLP2, of their shed ectodomains, and of their membrane-bound C-terminal fragments. In particular, APP processing appears to be tightly regulated, with changes in beta-cleaved APPs (APPsbeta) being compensated for by changes in alpha-cleaved APPs (APPsalpha). In contrast, the total levels of soluble cleaved APLP1 and APLP2 species were less tightly regulated, and fluctuated with BACE1 expression. Importantly, the production of ICDs for all three proteins was not decreased by loss of BACE1 activity. These results indicate that BACE1 is involved in regulating ectodomain shedding, maturation and trafficking of the APP family of proteins. Consequently, whereas inhibition of BACE1 is unlikely to adversely affect potential ICD-mediated signaling, it may alter other important facets of amyloid precursor-like protein/APP biology.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/physiology , Aspartic Acid Endopeptidases/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Gene Deletion , Gene Expression Regulation, Enzymologic , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Structure, Tertiary
14.
Brain Res ; 1262: 89-99, 2009 Mar 25.
Article in English | MEDLINE | ID: mdl-19401174

ABSTRACT

The amyloid precursor-like protein-1 (APLP1) is a member of a protein family that includes the Alzheimer's disease-associated amyloid precursor protein (APP). While much is known about the proteolytic processing of APP, fewer details are available about APLP1. Using Chinese hamster ovarian cells stably transfected with human APLP1 and a novel juxtamembrane anti-APLP1 antibody, we demonstrate the detection of a secreted approximately 3.5 kDa APLP1-derived peptide (ALP-1). The production of this peptide is abolished by inhibition of gamma-secretase, but not beta-secretase, suggesting that ALP-1 is analogous to the p3 fragment produced from APP. However, unlike p3 or Abeta, ALP-1 shows no obvious propensity for aggregation and is not toxic to neuronal cells. Moreover, using two distinct experimental paradigms, we demonstrate that neither cell-derived nor chemically synthesized ALP-1 influences the oligomerization or aggregation of Abeta.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Protein Precursor/immunology , Animals , Antibodies/pharmacology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Culture Media, Conditioned/pharmacology , Hippocampus/cytology , Humans , Molecular Sequence Data , Neurons/cytology , Neurons/physiology , Neurotoxins/genetics , Neurotoxins/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/genetics , Peptide Fragments/metabolism , Rabbits , Rats , Rats, Wistar , Transfection
15.
FEBS J ; 276(5): 1266-81, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19175671

ABSTRACT

We report the development of a high-level bacterial expression system for the Alzheimer's disease-associated amyloid beta-peptide (Abeta), together with a scaleable and inexpensive purification procedure. Abeta(1-40) and Abeta(1-42) coding sequences together with added ATG codons were cloned directly into a Pet vector to facilitate production of Met-Abeta(1-40) and Met-Abeta(1-42), referred to as Abeta(M1-40) and Abeta(M1-42), respectively. The expression sequences were designed using codons preferred by Escherichia coli, and the two peptides were expressed in this host in inclusion bodies. Peptides were purified from inclusion bodies using a combination of anion-exchange chromatography and centrifugal filtration. The method described requires little specialized equipment and provides a facile and inexpensive procedure for production of large amounts of very pure Abeta peptides. Recombinant peptides generated using this protocol produced amyloid fibrils that were indistinguishable from those formed by chemically synthesized Abeta1-40 and Abeta1-42. Formation of fibrils by all peptides was concentration-dependent, and exhibited kinetics typical of a nucleation-dependent polymerization reaction. Recombinant and synthetic peptides exhibited a similar toxic effect on hippocampal neurons, with acute treatment causing inhibition of MTT reduction, and chronic treatment resulting in neuritic degeneration and cell loss.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/isolation & purification , Peptide Fragments/isolation & purification , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Base Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Profiling , Immunohistochemistry , Kinetics , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/metabolism , Rats , Rats, Wistar
16.
J Am Chem Soc ; 130(46): 15437-43, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-18954050

ABSTRACT

Copolymeric NiPAM:BAM nanoparticles of varying hydrophobicity were found to retard fibrillation of the Alzheimer's disease-associated amyloid beta protein (Abeta). We found that these nanoparticles affect mainly the nucleation step of Abeta fibrillation. The elongation step is largely unaffected by the particles, and once the Abeta is nucleated, the fibrillation process occurs with the same rate as in the absence of nanoparticles. The extension of the lag phase for fibrillation of Abeta is strongly dependent on both the amount and surface character of the nanoparticles. Surface plasmon resonance studies show that Abeta binds to the nanoparticles and provide rate and equilibrium constants for the interaction. Numerical analysis of the kinetic data for fibrillation suggests that binding of monomeric Abeta and prefibrillar oligomers to the nanoparticles prevents fibrillation. Moreover, we find that fibrillation of Abeta initiated in the absence of nanoparticles can be reversed by addition of nanoparticles up to a particular time point before mature fibrils appear.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Nanoparticles/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Polymers/chemistry , Amyloid beta-Peptides/ultrastructure , Kinetics , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Peptide Fragments/ultrastructure , Protein Binding , Surface Plasmon Resonance , Time Factors
17.
J Neurochem ; 103(3): 914-26, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17711425

ABSTRACT

The age-related deficit in long-term potentiation (LTP) in the dentate gyrus is positively correlated with hippocampal concentration of the pro-inflammatory cytokine, interleukin-1beta (IL-1beta). Previous evidence also indicates that the inhibition of LTP induced by intracerebroventricular injection of amyloid-beta(1-40) (Abeta) is accompanied by increased hippocampal IL-1beta concentration and IL-1beta-stimulated signalling, specifically activation of the stress-activated protein kinase, c-jun N-terminal kinase (JNK). We considered that the underlying age-related neuroinflammation may render older rats more susceptible to Abeta administration and, to investigate this, young, middle-aged and aged rats were injected intracerebroventricularly with Abeta or vehicle. Hippocampal IL-1beta concentration, JNK phosphorylation, expression of the putative Abeta receptor, Receptor for advanced glycation end products (RAGE) and the microglial cell surface marker, CD40 were assessed. We report that Abeta inhibited LTP in a concentration-dependent manner in young rats and that this was accompanied by concentration-dependent increases in hippocampal IL-1beta and expression of phosphorylated JNK, RAGE and CD40. While 20 micromol/L Abeta exerted no significant effect on LTP in young rats, it inhibited LTP in middle-aged and aged rats and the increased vulnerability of aged rats was associated with increased IL-1beta concentration. Treatment of rats with eicosapentaenoic acid attenuated the inhibitory effect of 60 micromol/L Abeta on LTP in young rats and the effect of 20 micromol/L Abeta in middle-aged and aged rats. We present evidence which indicates that the effect of eicosapentaenoic acid may be linked with its ability to stimulate activation of peroxisome proliferator-activated receptor gamma.


Subject(s)
Aging/drug effects , Amyloid beta-Peptides/antagonists & inhibitors , Eicosapentaenoic Acid/pharmacology , Encephalitis/drug therapy , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Age Factors , Aging/metabolism , Amyloid beta-Peptides/toxicity , Animals , Animals, Newborn , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , CD40 Antigens/drug effects , CD40 Antigens/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Eicosapentaenoic Acid/therapeutic use , Encephalitis/metabolism , Encephalitis/physiopathology , Hippocampus/metabolism , Hippocampus/physiopathology , Injections, Intraventricular , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Long-Term Potentiation/physiology , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phosphorylation/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products , Receptors, Immunologic/drug effects , Receptors, Immunologic/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
18.
Neurobiol Aging ; 28(6): 845-55, 2007 Jun.
Article in English | MEDLINE | ID: mdl-16714069

ABSTRACT

Among the changes that occur in the hippocampus with age, is a deficit in long-term potentiation (LTP). This impairment is associated with inflammatory changes, which are typified by increased concentration of the pro-inflammatory cytokine interleukin-1beta (IL-1beta). Activated microglia are the most likely cell source of IL-1beta, but data demonstrating an age-related increase in microglial activation is equivocal. Here we demonstrate that the age-related deficit in LTP is accompanied by increased expression of cell surface markers of activated microglia (major histocompatibility complex II and CD40) and increased IL-1beta production, and that these changes may be stimulated by interferon-gamma. Treatment of aged rats with eicosapentaenoic acid (EPA) attenuates these changes and we suggest that IL-4 mediates the action of EPA. We demonstrate that aged rats exhibit an exaggerated response to intracerebroventricular injection of beta-amyloid peptide 1-40 (Abeta). Thus Abeta inhibited LTP in aged, but not young, rats and induced a further increase in hippocampal IL-1beta concentration. Of particular significance is the demonstration that EPA protects the aged brain so that the increased vulnerability to Abeta is ameliorated in EPA-treated rats.


Subject(s)
Aging , Amyloid beta-Peptides/administration & dosage , Eicosapentaenoic Acid/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Neuroprotective Agents/pharmacology , Peptide Fragments/administration & dosage , Analysis of Variance , Animals , Animals, Newborn , CD40 Antigens/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Histocompatibility Antigens Class II/metabolism , Injections, Intraventricular , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Neuroglia/drug effects , Nitric Oxide/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Wistar
19.
J Biol Chem ; 278(30): 27971-80, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12738769

ABSTRACT

Amyloid-beta (Abeta) is a major constituent of the neuritic plaque found in the brain of Alzheimer's disease patients, and a great deal of evidence suggests that the neuronal loss that is associated with the disease is a consequence of the actions of Abeta. In the past few years, it has become apparent that activation of c-Jun N-terminal kinase (JNK) mediates some of the effects of Abeta on cultured cells; in particular, the evidence suggests that Abeta-triggered JNK activation leads to cell death. In this study, we investigated the effect of intracerebroventricular injection of Abeta(1-40) on signaling events in the hippocampus and on long term potentiation in Schaffer collateral CA1 pyramidal cell synapses in vivo. We report that Abeta(1-40) induced activation of JNK in CA1 and that this was coupled with expression of the proapoptotic protein, Bax, cytosolic cytochrome c, poly-(ADP-ribose) polymerase cleavage, and Fas ligand expression in the hippocampus. These data indicate that Abeta(1-40) inhibited expression of long term potentiation, and this effect was abrogated by administration of the JNK inhibitor peptide, D-JNKI1. In parallel with these findings, we observed that Abeta-induced changes in caspase-3 activation and TdT-mediated dUTP nick-end labeling staining in neuronal cultured cells were inhibited by D-JNKI1. We present evidence suggesting that interleukin (IL)-1beta plays a significant role in mediating the effects of Abeta(1-40) because Abeta(1-40) increased hippocampal IL-1beta and because several effects of Abeta(1-40) were inhibited by the caspase-1 inhibitor Ac-YVAD-CMK. On the basis of our findings, we propose that Abeta-induced changes in hippocampal plasticity are likely to be dependent upon IL-1beta-triggered activation of JNK.


Subject(s)
Amyloid beta-Peptides/chemistry , Hippocampus/metabolism , Mitogen-Activated Protein Kinases/metabolism , Peptide Fragments/chemistry , Proto-Oncogene Proteins c-bcl-2 , Signal Transduction , Animals , Caspase 3 , Caspases/metabolism , Cell Death , Cytochrome c Group/biosynthesis , Cytosol/metabolism , DNA Fragmentation , Enzyme Activation , Fas Ligand Protein , Immunohistochemistry , In Situ Nick-End Labeling , Interleukin-1/metabolism , Interleukin-1/physiology , JNK Mitogen-Activated Protein Kinases , Long-Term Potentiation , Male , Membrane Glycoproteins/metabolism , Neurons/metabolism , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...