Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Neurosci ; 43(50): 8812-8824, 2023 12 13.
Article in English | MEDLINE | ID: mdl-37884349

ABSTRACT

Accumulation of amyloid-ß peptide (Aß) aggregates in synapses may contribute to the profound synaptic loss characteristic of Alzheimer's disease (AD). The origin of synaptic Aß aggregates remains elusive, but loss of endosomal proteostasis may trigger their formation. In this study, we identified the synaptic compartments where Aß accumulates, and performed a longitudinal analysis of synaptosomes isolated from brains of TgCRND8 APP transgenic mice of either sex. To evaluate the specific contribution of Aß-degrading protease endothelin-converting enzyme (ECE-1) to synaptic/endosomal Aß homeostasis, we analyzed the effect of partial Ece1 KO in brain and complete ECE1 KO in SH-SY5Y cells. Global inhibition of ECE family members was used to further assess their role in preventing synaptic Aß accumulation. Results showed that, before extracellular amyloid deposition, synapses were burdened with detergent-soluble Aß monomers, oligomers, and fibrils. Levels of all soluble Aß species declined thereafter, as Aß42 turned progressively insoluble and accumulated in Aß-producing synaptic endosomal vesicles with characteristics of multivesicular bodies. Accordingly, fibrillar Aß was detected in brain exosomes. ECE-1-deficient mice had significantly increased endogenous synaptosomal Aß42 levels, and protease inhibitor experiments showed that, in TgCRND8 mice, synaptic Aß42 became nearly resistant to degradation by ECE-related proteases. Our study supports that Aß accumulating in synapses is produced locally, within endosomes, and does not require the presence of amyloid plaques. ECE-1 is a determinant factor controlling the accumulation and fibrillization of nascent Aß in endosomes and, in TgCRND8 mice, Aß overproduction causes rapid loss of Aß42 solubility that curtails ECE-mediated degradation.SIGNIFICANCE STATEMENT Deposition of aggregated Aß in extracellular plaques is a defining feature of AD. Aß aggregates also accumulate in synapses and may contribute to the profound synaptic loss and cognitive dysfunction typical of the disease. However, it is not clear whether synaptotoxic Aß is mainly derived from plaques or if it is produced and aggregated locally, within affected synaptic compartments. Filling this knowledge gap is important for the development of an effective treatment for AD, as extracellular and intrasynaptic pools of Aß may not be equally modulated by immunotherapies or other therapeutic approaches. In this manuscript, we provide evidence that Aß aggregates building up in synapses are formed locally, within synaptic endosomes, because of disruptions in nascent Aß proteostasis.


Subject(s)
Alzheimer Disease , Amyloidosis , Neuroblastoma , Humans , Mice , Animals , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Neurons/metabolism , Neuroblastoma/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Endosomes/metabolism , Plaque, Amyloid/metabolism
2.
J Neurochem ; 165(6): 860-873, 2023 06.
Article in English | MEDLINE | ID: mdl-37002186

ABSTRACT

Aggregation and accumulation of amyloid-ß peptide (Aß) are a critical trigger for the onset of Alzheimer's disease (AD). While the plaques are the most outstanding Aß pathological feature, much of the recent research emphasis has been on soluble Aß species because of their diffusible, proinflammatory, and toxic properties. The focus on soluble aggregated Aß species has also increased the interest in antibodies that are selective for different Aß conformations. In the current study, we developed and characterized a new class of monoclonal antibodies (referred to as mAbSL) that are selective for Aß protofibrils. Cloning and sequencing of the heavy and light chain variable regions for multiple antibodies identified sequence characteristics that may impart the conformational selectivity by the antibodies. Transfection of FreeStyle 293F cells with the plasmids permitted in-house expression and purification of mAbSL antibodies along with non-conformation-selective Aß monoclonal antibodies (Aß mAbs). Several of the purified mAbSL antibodies demonstrated significant affinity and selectivity for Aß42 protofibrils compared with Aß42 monomers and Aß42 fibrils. Competition ELISA assays assessing the best overall antibody, mAbSL 113, yielded affinity constants of 7 nM for the antibody-Aß42 protofibril interaction, while the affinity for either Aß42 monomers or Aß42 fibrils was roughly 80 times higher. mAbSL 113 significantly inhibited Aß42 monomer aggregation by a unique mechanism compared with the inhibition displayed by Aß mAb 513. Aß42 protofibril dynamics were also markedly altered in the presence of mAbSL 113, whereby insoluble complex formation and protofibril deposition were stimulated by the antibody at low substoichiometric molar ratios. As the field contemplates the therapeutic effectiveness of Aß conformation-selective antibodies, the findings presented here demonstrate new information on a monoclonal antibody that selectively targets Aß protofibrils and impacts Aß dynamics.


Subject(s)
Alzheimer Disease , Antibodies, Monoclonal , Humans , Peptide Fragments/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Enzyme-Linked Immunosorbent Assay
3.
Biochim Biophys Acta Proteins Proteom ; 1870(9): 140817, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35905824

ABSTRACT

Three decades of research, both in vitro and in vivo, have demonstrated the conformational heterogeneity that is displayed by the amyloid ß peptide (Aß) in Alzheimer's disease (AD). Understanding the distinct properties between Aß conformations and how conformation may impact cellular activity remain open questions, yet still continue to provide new insights into protein misfolding and aggregation. In particular, there is interest in the group of soluble oligomeric prefibrillar Aß species comprising lower molecular weight oligomers up to larger protofibrils. In the current study, a number of strategies were utilized to separate Aß protofibrils and oligomers and show that the smaller Aß oligomers have a much different conformation than Aß protofibrils. The differences were consistent for both Aß40 and Aß42. Protofibrils bound thioflavin T to a greater extent than oligomers, and were highly enriched in ß-sheet secondary structure. Aß oligomers possessed a more open structure with significant solvent exposure of hydrophobic domains as determined by tryptophan fluorescence and bis-ANS binding, respectively. The protofibril-selective antibody AbSL readily discerned conformational differences between protofibrils and oligomers. The more developed structure for Aß protofibrils ultimately proved critical for provoking the release of tumor necrosis factor α from microglial cells. The findings demonstrated a dependency on ß-sheet structure for soluble Aß aggregates to cause a microglial inflammatory response. The Aß aggregation process yields many conformationally-varied species with different levels of ß-structure and exposed hydrophobicity. The conformation elements likely determine biological activity and pathogenicity.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Peptide Fragments/chemistry , Protein Conformation, beta-Strand
4.
ACS Infect Dis ; 8(6): 1171-1178, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35612826

ABSTRACT

Sepsis is a serious medical condition characterized by bacterial infection and a subsequent massive systemic inflammatory response. In an effort to identify compounds that block lipopolysaccharide (LPS)-induced inflammation reported herein is the development of simple Lipid-A analogues that lack a disaccharide core yet still possess potent antagonistic activity against LPS. The structure of the new lead compound was developed based on predictive computational experiments. LPS antagonism by the lead compound was not straightforward, and a biphasic effect was observed suggesting a possibility of more than one binding site. An IC50 value of 13 nM for the new compound was determined for the possible high affinity site. The combination of computational, synthetic, and biological studies revealed new structural determinants of these simplified analogues. It is expected that the acquired information will aid future design of LPS targeting glycopharmaceuticals.


Subject(s)
Lipid A , Lipopolysaccharides , Binding Sites , Humans , Inflammation , Lipid A/chemistry , Lipopolysaccharides/chemistry , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/metabolism
5.
Biotechnol Prog ; 37(4): e3153, 2021 07.
Article in English | MEDLINE | ID: mdl-33811748

ABSTRACT

The NLRP3 inflammasome is a key intracellular component of the innate immune response. It is a three-protein complex essential for the production of mature interleukin 1-ß. The complex, which is comprised of three proteins, NLRP3, ASC, and pro-caspase-1, has been implicated in the physiological response to pathogenic elements of cardiovascular disease and Alzheimer's disease. Investigations into the properties of the three proteins can be aided by larger-scale recombinant expression to produce adequate amounts. In the current study, a variety of NLRP3 inflammasome proteins were expressed in the ExpiCHO-S mammalian cell system with a particular focus on ASC. ASC fusion proteins with glutathione-S transferase, maltose-binding protein, and SUMO increased solubility and aided in determining the stability and oligomerization propensity of individual ASC domains and full-length ASC. ASC oligomerization was highly sensitive to protein concentration, ionic strength, and mutation. These observations provided strategic ways to enhance protein purification and characterize ASC oligomerization. The ExpiCHO-S expression system consistently produced high-yield recombinant NLRP3 inflammasome proteins which led to a further understanding of ASC oligomerization.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Inflammasomes/genetics , Inflammasomes/metabolism , Interleukin-1beta , Mammals/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
6.
World J Biol Chem ; 12(1): 1-14, 2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33552397

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs), including MMP-9, are an integral part of the immune response and are upregulated in response to a variety of stimuli. New details continue to emerge concerning the mechanistic and regulatory pathways that mediate MMP-9 secretion. There is significant evidence for regulation of inflammation by dimethyl sulfoxide (DMSO) and 3',5'-cyclic adenosine monophosphate (cAMP), thus investigation of how these two molecules may regulate both MMP-9 and tumor necrosis factor α (TNFα) secretion by human monocytes was of high interest. The hypothesis tested in this study was that DMSO and cAMP regulate MMP-9 and TNFα secretion by distinct mechanisms. AIM: To investigate the regulation of lipopolysaccharide (LPS)-stimulated MMP-9 and tumor necrosis factor α secretion in THP-1 human monocytes by dimethyl sulfoxide and cAMP. METHODS: The paper describes a basic research study using THP-1 human monocyte cells. All experiments were conducted at the University of Missouri-St. Louis in the Department of Chemistry and Biochemistry. Human monocyte cells were grown, cultured, and prepared for experiments in the University of Missouri-St. Louis Cell Culture Facility as per accepted guidelines. Cells were treated with LPS for selected exposure times and the conditioned medium was collected for analysis of MMP-9 and TNFα production. Inhibitors including DMSO, cAMP regulators, and anti-TNFα antibody were added to the cells prior to LPS treatment. MMP-9 secretion was analyzed by gel electrophoresis/western blot and quantitated by ImageJ software. TNFα secretion was analyzed by enzyme-linked immuno sorbent assay. All data is presented as the average and standard error for at least 3 trials. Statistical analysis was done using a two-tailed paired Student t-test. P values less than 0.05 were considered significant and designated as such in the Figures. LPS and cAMP regulators were from Sigma-Aldrich, MMP-9 standard and antibody and TNFα antibodies were from R&D Systems, and amyloid-ß peptide was from rPeptide. RESULTS: In our investigation of MMP-9 secretion from THP-1 human monocytes, we made the following findings. Inclusion of DMSO in the cell treatment inhibited LPS-induced MMP-9, but not TNFα, secretion. Inclusion of DMSO in the cell treatment at different concentrations inhibited LPS-induced MMP-9 secretion in a dose-dependent fashion. A cell-permeable cAMP analog, dibutyryl cAMP, inhibited both LPS-induced MMP-9 and TNFα secretion. Pretreatment of the cells with the adenylyl cyclase activator forskolin inhibited LPS-induced MMP-9 and TNFα secretion. Pretreatment of the cells with the general cAMP phosphodiesterase inhibitor IBMX reduced LPS-induced MMP-9 and TNFα in a dose-dependent fashion. Pre-treatment of monocytes with an anti-TNFα antibody blocked LPS-induced MMP-9 and TNFα secretion. Amyloid-ß peptide induced MMP-9 secretion, which occurred much later than TNFα secretion. The latter two findings strongly suggested an upstream role for TNFα in mediating LPS-stimulate MMP-9 secretion. CONCLUSION: The cumulative data indicated that MMP-9 secretion was a distinct process from TNFα secretion and occurred downstream. First, DMSO inhibited MMP-9, but not TNFα, suggesting that the MMP-9 secretion process was selectively altered. Second, cAMP inhibited both MMP-9 and TNFα with a similar potency, but at different monocyte cell exposure time points. The pattern of cAMP inhibition for these two molecules suggested that MMP-9 secretion lies downstream of TNFα and that TNFα may a key component of the pathway leading to MMP-9 secretion. This temporal relationship fit a model whereby early TNFα secretion directly led to later MMP-9 secretion. Lastly, antibody-blocking of TNFα diminished MMP-9 secretion, suggesting a direct link between TNFα secretion and MMP-9 secretion.

7.
Arch Biochem Biophys ; 699: 108753, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33453207

ABSTRACT

This review takes a closer look at the structural components of the molecules involved in the processes leading to caspase-1 activation. Interleukins 1ß and 18 (IL-1ß, IL-18) are well-known proinflammatory cytokines that are produced following cleavage of their respective precursor proteins by the cysteine protease caspase-1. Active caspase-1 is the final step of the NLRP3 inflammasome, a three-protein intracellular complex involved in inflammation and induction of pyroptosis (a proinflammatory cell-death process). NLRP3 activators facilitate assembly of the inflammasome complex and subsequent activation of caspase-1 by autoproteolysis. However, the definitive structural components of active caspase-1 are still unclear and new data add to the complexity of this process. This review outlines the historical and recent findings that provide supporting evidence for the structural aspects of caspase-1 autoproteolysis and activation.


Subject(s)
Caspase 1/metabolism , Animals , Caspase 1/chemistry , Cell Line, Tumor , Enzyme Activation/physiology , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Multimerization/physiology , Proteolysis
9.
J Neurochem ; 153(1): 7-9, 2020 04.
Article in English | MEDLINE | ID: mdl-32037541

ABSTRACT

Protein aggregation plays a central role in numerous neurodegenerative diseases. The key proteins in these diseases are of significant importance, but their investigation can be challenging due to unique properties of protein misfolding and oligomerization. Alpha-synuclein protein (α-Syn) is the predominant component of Lewy Bodies in Parkinson's disease (PD) and is a member of this class of proteins. Many α-Syn studies are limited by the inability to separate various monomeric, oligomeric, and fibrillar forms of the protein from heterogeneous mixtures. This Editorial Highlight summarizes the impact of a study published in the current issue of Journal of Neurochemistry, in which Lashuel and colleagues developed a simple, rapid centrifugation- and filter-based method for separating, isolating, and quantifying different forms of α-Syn. The researchers used electron microscopy, SDS-PAGE, circular dichroism, and protein assays to carefully validate the method and quantitate α-Syn yields and loss. The publication of this new method will not only aid in future studies of α-Syn, but will likely extend to other proteins that underlie a variety of neurodegenerative diseases.


Subject(s)
Centrifugation/methods , Filtration/methods , alpha-Synuclein/isolation & purification , Humans , Parkinson Disease , Protein Aggregation, Pathological , Reproducibility of Results , alpha-Synuclein/analysis , alpha-Synuclein/chemistry
10.
Nat Med ; 26(1): 131-142, 2020 01.
Article in English | MEDLINE | ID: mdl-31932797

ABSTRACT

Glia have been implicated in Alzheimer's disease (AD) pathogenesis. Variants of the microglia receptor triggering receptor expressed on myeloid cells 2 (TREM2) increase AD risk, and activation of disease-associated microglia (DAM) is dependent on TREM2 in mouse models of AD. We surveyed gene-expression changes associated with AD pathology and TREM2 in 5XFAD mice and in human AD by single-nucleus RNA sequencing. We confirmed the presence of Trem2-dependent DAM and identified a previously undiscovered Serpina3n+C4b+ reactive oligodendrocyte population in mice. Interestingly, remarkably different glial phenotypes were evident in human AD. Microglia signature was reminiscent of IRF8-driven reactive microglia in peripheral-nerve injury. Oligodendrocyte signatures suggested impaired axonal myelination and metabolic adaptation to neuronal degeneration. Astrocyte profiles indicated weakened metabolic coordination with neurons. Notably, the reactive phenotype of microglia was less evident in TREM2-R47H and TREM2-R62H carriers than in non-carriers, demonstrating a TREM2 requirement in both mouse and human AD, despite the marked species-specific differences.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Transcriptome/genetics , Aged , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Axons/pathology , Brain/metabolism , Brain/pathology , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Middle Aged , Nerve Degeneration/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Transcription, Genetic
11.
J Neurochem ; 149(5): 562-581, 2019 06.
Article in English | MEDLINE | ID: mdl-30702751

ABSTRACT

This review discusses the profound connection between microglia, neuroinflammation, and Alzheimer's disease (AD). Theories have been postulated, tested, and modified over several decades. The findings have further bolstered the belief that microglia-mediated inflammation is both a product and contributor to AD pathology and progression. Distinct microglia phenotypes and their function, microglial recognition and response to protein aggregates in AD, and the overall role of microglia in AD are areas that have received considerable research attention and yielded significant results. The following article provides a historical perspective of microglia, a detailed discussion of multiple microglia phenotypes including dark microglia, and a review of a number of areas where microglia intersect with AD and other pathological neurological processes. The overall breadth of important discoveries achieved in these areas significantly strengthens the hypothesis that neuroinflammation plays a key role in AD. Future determination of the exact mechanisms by which microglia respond to, and attempt to mitigate, protein aggregation in AD may lead to new therapeutic strategies.


Subject(s)
Alzheimer Disease/immunology , Inflammation/immunology , Microglia/immunology , Nerve Degeneration/immunology , Alzheimer Disease/pathology , Animals , Humans , Microglia/metabolism , Nerve Degeneration/pathology
12.
ACS Chem Neurosci ; 9(6): 1416-1425, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29543435

ABSTRACT

Microvesicles (MVs) and exosomes comprise a class of cell-secreted particles termed extracellular vesicles (EVs). These cargo-holding vesicles mediate cell-to-cell communication and have recently been implicated in neurodegenerative diseases such as Alzheimer's disease (AD). The two types of EVs are distinguished by the mechanism of cell release and their size, with the smaller exosomes and the larger MVs ranging from 30 to 100 nm and 100 nm to 1 µm in diameter, respectively. MV numbers are increased in AD and appear to interact with amyloid-ß peptide (Aß), the primary protein component of the neuritic plaques in the AD brain. Because microglial cells play such an important role in AD-linked neuroinflammation, we sought to characterize MVs shed from microglial cells, better understand MV interactions with Aß, and determine whether internalized Aß may be incorporated into secreted MVs. Multiple strategies were used to characterize MVs shed from BV-2 microglia after ATP stimulation. Confocal images of isolated MVs bound to fluorescently labeled annexin-V via externalized phosphatidylserine revealed a polydisperse population of small spherical structures. Dynamic light scattering measurements yielded MV diameters ranging from 150 to 600 nm. Electron microscopy of resin-embedded MVs cut into thin slices showed well-defined uranyl acetate-stained ring-like structures in a similar diameter range. The use of a fluorescently labeled membrane insertion probe, NBD C6-HPC, effectively tracked MVs in binding experiments, and an Aß ELISA confirmed a strong interaction between MVs and Aß protofibrils but not Aß monomers. Despite the lesser monomer interaction, MVs had an inhibitory effect on monomer aggregation. Primary microglia rapidly internalized Aß protofibrils, and subsequent stimulation of the microglia with ATP resulted in the release of MVs containing the internalized Aß protofibrils. The role of MVs in neurodegeneration and inflammation is an emerging area, and further knowledge of MV interaction with Aß may shed light on extracellular spread and influence on neurotoxicity and neuroinflammation.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Extracellular Vesicles/metabolism , Microglia/metabolism , Animals , Brain/metabolism , Cell Movement/physiology , Inflammation/metabolism , Mice , Microglia/drug effects , Peptide Fragments/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
J Neurochem ; 143(6): 736-749, 2017 12.
Article in English | MEDLINE | ID: mdl-28881033

ABSTRACT

Aggregation and accumulation of amyloid-ß peptide (Aß) is a key component of Alzheimer's disease (AD). While monomeric Aß appears to be benign, oligomers adopt a biologically detrimental structure. These soluble structures can be detected in AD brain tissue by antibodies that demonstrate selectivity for aggregated Aß. Protofibrils are a subset of soluble oligomeric Aß species and are described as small (< 100 nm) curvilinear assemblies enriched in ß-sheet structure. Our own in vitro studies demonstrate that microglial cells are much more sensitive to soluble Aß42 protofibrils compared to Aß42 monomer or insoluble Aß42 fibrils. Protofibrils interact with microglia, trigger Toll-like receptor signaling, elicit cytokine transcription and expression, and are rapidly taken up by the cells. Because of the importance of this Aß species, we sought to develop an antibody that selectively recognizes protofibrils over other Aß species. Immunization of rabbits with isolated Aß42 protofibrils generated a high-titer anti serum with a strong affinity for Aß42 protofibrils. The antiserum, termed AbSL, was selective for Aß42 protofibrils over Aß42 monomers and Aß42 fibrils. AbSL did not react with amyloid precursor protein and recognized distinct pathological features in AD transgenic mouse brain slices. Competition studies with an Aß antibody that targets residues 1-16 indicated that the conformational epitope for AbSL involved the N-terminal region of protofibrils in some manner. The newly developed antibody may have potential diagnostic and therapeutic uses in AD tissue and patients, and targeting of protofibrils in AD may have beneficial effects. Read the Editorial Highlight for this article on page 621. Cover Image for this issue: doi. 10.1111/jnc.13827.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Antibodies/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Animals , Antibody Specificity , Epitopes , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Conformation, beta-Strand
14.
Brain Res ; 1648(Pt A): 485-495, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27531183

ABSTRACT

One pathological hallmark of Alzheimer's disease (AD) is the accumulation of amyloid-ß peptide (Aß) in the affected brain. While there are numerous deleterious effects of Aß accumulation, there is general agreement that a sustained inflammatory response to aggregated Aß contributes to progressive neurodegeneration in AD and microglial cells play a significant role in this process. Our laboratory and others have shown that small soluble aggregates of Aß activate a microglia-mediated inflammatory response. One component of the response involves internalization of extracellular Aß, and this process is likely very sensitive to Aß structure. In this study we analyzed the proclivity of microglia for internalization of Aß42 monomers and protofibrils using fluorescently-labeled Aß. Both Aß42 species were labeled directly via amino linkage with an Alexa Fluor 488 tetrafluorophenyl ester (AF488-TFP) and then isolated individually by chromatography. Aß42 protofibrils retained their size and morphological properties after labeling but monomers had a much higher stoichiometry of labeling compared to protofibrils. Primary murine microglia internalized AF488-Aß42 protofibrils rapidly and in significant amounts compared to AF488-Aß42 monomers. Microglial internalization of protofibrils was dependent on time and concentration, and corresponded with tumor necrosis factor α secretion. In competition studies, unlabeled Aß42 protofibril internalization, detected by immunostaining, did not diminish AF488-protofibril uptake. Internalized AF488-Aß42 protofibrils were found widely dispersed in the cytosol with some lysosomal accumulation but little degradation. These studies highlight the sensitivity that microglia exhibit to Aß structure in the internalization process and emphasize their affinity for soluble Aß protofibrils.


Subject(s)
Amyloid beta-Peptides/metabolism , Microglia/metabolism , Peptide Fragments/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/ultrastructure , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Tumor Necrosis Factor-alpha/metabolism
15.
J Neurosci ; 36(32): 8471-86, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27511018

ABSTRACT

UNLABELLED: Prior work suggests that amyloid precursor protein (APP) can function as a proinflammatory receptor on immune cells, such as monocytes and microglia. Therefore, we hypothesized that APP serves this function in microglia during Alzheimer's disease. Although fibrillar amyloid ß (Aß)-stimulated cytokine secretion from both wild-type and APP knock-out (mAPP(-/-)) microglial cultures, oligomeric Aß was unable to stimulate increased secretion from mAPP(-/-) cells. This was consistent with an ability of oligomeric Aß to bind APP. Similarly, intracerebroventricular infusions of oligomeric Aß produced less microgliosis in mAPP(-/-) mice compared with wild-type mice. The mAPP(-/-) mice crossed to an APP/PS1 transgenic mouse line demonstrated reduced microgliosis and cytokine levels and improved memory compared with wild-type mice despite robust fibrillar Aß plaque deposition. These data define a novel function for microglial APP in regulating their ability to acquire a proinflammatory phenotype during disease. SIGNIFICANCE STATEMENT: A hallmark of Alzheimer's disease (AD) brains is the accumulation of amyloid ß (Aß) peptide within plaques robustly invested with reactive microglia. This supports the notion that Aß stimulation of microglial activation is one source of brain inflammatory changes during disease. Aß is a cleavage product of the ubiquitously expressed amyloid precursor protein (APP) and is able to self-associate into a wide variety of differently sized and structurally distinct multimers. In this study, we demonstrate both in vitro and in vivo that nonfibrillar, oligomeric forms of Aß are able to interact with the parent APP protein to stimulate microglial activation. This provides a mechanism by which metabolism of APP results in possible autocrine or paracrine Aß production to drive the microgliosis associated with AD brains.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Microglia/metabolism , Adaptation, Ocular/genetics , Adaptation, Ocular/physiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/pharmacology , Animals , Astrocytes/metabolism , Cell Proliferation/genetics , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Exploratory Behavior/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morpholinos/pharmacology , Mutation/genetics , Phenotype , Presenilin-1/genetics , Presenilin-1/metabolism
16.
Arch Biochem Biophys ; 597: 1-11, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27013205

ABSTRACT

Recent findings suggest that the senile plaques in Alzheimer's disease may contain soluble amyloid-ß peptide (Aß) fibril precursors along with insoluble fibrils. These soluble Aß species, including oligomers and protofibrils, have been well-studied in vitro and are formed via non-covalent self-assembly of Aß monomers. While both 40- and 42-residue forms of Aß are observed in the human body, the majority of the Aß aggregation work has been conducted on Aß42 or Aß40 separately, with relatively few investigations of mixtures. In order to study the effect of different combinations of Aß40 and Aß42 on protofibril formation, mixtures of either dry solid peptide, or purified Aß40 and Aß42 monomer solutions were mixed together and protofibril/monomer distributions were quantified. Increases in the Aß42/Aß40 ratio increased protofibril formation but the presence of Aß40 in the mixed Aß solutions had a significant negative impact on protofibril formation compared to equivalent solutions of pure Aß42. Protofibril size was less affected, but ß-sheet structure increased with protofibrils formed from higher Aß42/Aß40 ratio solutions. Direct measurement of Aß42/Aß40 ratios by C-terminal-selective ELISA found very little Aß40 incorporated into protofibrils. The cumulative data emphasizes the critical importance of Aß42, yet establishes Aß40 as a regulator of Aß42 aggregation.


Subject(s)
Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Protein Aggregates , Humans , Protein Structure, Secondary
17.
Biochemistry ; 54(13): 2193-204, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25756466

ABSTRACT

Some of the pathological hallmarks of the Alzheimer's disease brain are senile plaques composed of insoluble amyloid-ß protein (Aß) fibrils. However, much of the recent emphasis in research has been on soluble Aß aggregates in response to a growing body of evidence that shows that these species may be more neurotoxic than fibrils. Within this subset of soluble aggregated Aß are protofibrils and oligomers. Although each species has been widely investigated separately, few studies have directly compared and contrasted their physical properties. In this work, we examined well-recognized preparations of Aß(1-42) oligomers and protofibrils with multiangle (MALS) and dynamic (DLS) light scattering in line with, or following, size-exclusion chromatography (SEC). Multiple SEC-MALS analyses of protofibrils revealed molecular weight (Mw) gradients ranging from 200 to 2600 kDa. Oligomeric Aß species are generally considered to be a smaller and more nascent than protofibrils. However, oligomer Mw values ranged from 225 to 3000 kDa, larger than that for protofibrils. Root-mean-square radius (Rg) values correlated with the Mw trends with protofibril Rg values ranging from 16 to 35 nm, while oligomers produced one population at 40-43 nm with a more disperse population from 22 to 39 nm. Hydrodynamic radius (RH) measurements by DLS and thioflavin T fluorescence measurements indicated that protofibrils and oligomers had commonalities, yet electron microscopy revealed morphological differences between the two. SEC-purified Aß(1-42) monomer at lower concentrations was slower to nucleate but formed protofibrils (1500 kDa) or soluble protofilaments (3000 kDa) depending on the buffer type. The findings from these studies shed new light on the similarities and differences between distinct soluble aggregated Aß species.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Peptide Fragments/chemistry , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/isolation & purification , Benzothiazoles , Chromatography, Gel , Circular Dichroism , Microscopy, Electron , Peptide Fragments/analysis , Peptide Fragments/isolation & purification , Protein Structure, Secondary , Scattering, Radiation , Solubility , Spectrometry, Fluorescence , Thiazoles/chemistry
18.
Biochem Biophys Res Commun ; 454(1): 239-44, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25451248

ABSTRACT

Neuroinflammation triggered by accumulation of amyloid-ß protein (Aß) is a significant component of the Alzheimer's disease (AD) brain. Senile plaques composed of Aß attract and activate microglia cells resulting in cytokine secretion and a proinflammatory environment. The mechanism by which Aß activates microglia is complex and involves numerous cellular components. One receptor potentially involved in Aß recognition and the ensuing microglia proinflammatory response is CD47. Since there is significant interest in soluble aggregated Aß species, we sought to determine if CD47 plays a key role in microglia cytokine release stimulated by soluble Aß(1-42) protofibrils. Pretreatment of primary murine microglia with the CD47 antagonist peptide 4N1K significantly and potently inhibited both tumor necrosis factor-α (TNFα) and interleukin-1ß (IL-1ß) secretion stimulated by Aß(1-42) protofibrils. 4N1K displayed toxicity to the microglia but only at concentrations much higher than the observed inhibition. Surprisingly, 4N1K also potently inhibited TNFα secretion triggered by lipopolysaccharide which is not known to signal through CD47. Treatment of the microglia with a neutralizing anti-CD47 antibody failed to block the Aß protofibril response even though comparable samples were completely inhibited by 4N1K. Finally, Aß(1-42) protofibrils stimulated similar levels of secreted TNFα production in both wild-type and CD47(-/-) microglia and 4N1K still potently inhibited the Aß protofibril response even in the CD47(-/-) microglia. The overall findings demonstrated that the microglial proinflammatory response to Aß(1-42) protofibril is not dependent on CD47 and that 4N1K exhibits CD47-independent inhibitory activity.


Subject(s)
Amyloid beta-Peptides/metabolism , CD47 Antigen/metabolism , Microglia/immunology , Microglia/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid/chemistry , Amyloid/metabolism , Amyloid beta-Peptides/chemistry , Animals , Antibodies, Neutralizing/administration & dosage , CD47 Antigen/genetics , Cells, Cultured , Disease Models, Animal , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Oligopeptides/pharmacology , Peptide Fragments/chemistry , Tumor Necrosis Factor-alpha/metabolism
19.
Biochim Biophys Acta ; 1842(11): 2276-85, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25125050

ABSTRACT

Neuroinflammation is a characteristic feature of the Alzheimer's disease (AD) brain. Significant inflammatory markers such as activated microglia and cytokines can be found surrounding the extracellular senile plaques predominantly composed of amyloid-ß protein (Aß). Several innate immune pathways, including Toll-like receptors (TLRs) and the NLRP3 inflammasome, have been implicated in AD inflammation. Aß plays a primary role in activating these pathways which likely contributes to the progressive neurodegeneration in AD. In order to better understand the complexities of this interaction we investigated the inflammatory response of primary microglia to Aß(1-42) protofibrils. Aß(1-42) protofibrils triggered a time- and MyD88-dependent process that produced tumor necrosis factor alpha (TNFα) and interleukin-1ß (IL-1ß) mRNA, and intracellular pro and mature forms of IL-1ß protein. The accumulation of both IL-1ß forms indicated that Aß(1-42) protofibrils were able to prime and activate the NLRP3 inflammasome. Surprisingly, Aß-induced accumulation of intracellular mature IL-1ß did not translate into greater IL-1ß secretion. Instead, we found that Aß elicited a quantized burst of secreted IL-1ß and this process occurred even prior to Aß priming of the microglia suggesting a basal level of either pro or mature IL-1ß in the cultured primary microglia. The IL-1ß secretion burst was rapid but not sustained, yet could be re-evoked with additional Aß stimulation. The findings from this study demonstrated multiple sites of IL-1ß regulation by Aß(1-42) protofibrils including TLR/MyD88-mediated priming, NLRP3 inflammasome activation, and modulation of the IL-1ß secretory process. These results underscore the wide-ranging effects of Aß on the innate immune response.

20.
Biomater Sci ; 2(1): 110-120, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-32481813

ABSTRACT

Microglial cells play a critical role in the propagation of neuroinflammation in the central nervous system. Microglia sense and respond to environmental signals including chemical, physical and biological cues from the surrounding cell/tissue components. In this project, our goal was to examine the effects of surface texture on BV-2 microglia morphology and function by comparing flat and nanoporous gold (np-Au) surfaces to the more conventional glass. The biocompatibility of np-Au with microglia was evaluated using functional cell assays and high resolution imaging with scanning electron microscopy (SEM). Microglia seeded on glass, ultra-flat gold (UF-Au), ultra-thin (UT) np-Au and np-Au monolith were adherent to all surfaces and their viability was not compromised as assessed by multiple toxicity assays. SEM revealed detailed morphological characteristics of adherent microglia and indicated few dramatic changes as a result of the different surfaces. Microglia proliferation was hampered by np-Au monolith but less by UT np-Au and not at all on UF-Au or glass. Microglial activation, measured by tumor necrosis factor α (TNFα) production, was fully functional (and equivalent) on all gold surfaces compared to glass. The present findings should help further the understanding of basic microglia biology on textured surfaces and more fully evaluate np-Au as a multi-functional biocompatible material. The knowledge obtained and technology developed will have a significant impact in the fabrication of nanoelectronic devices, chemical sensor development, porous nanostructured materials for BioMEMs/NEMs integration, and functional biomaterial coatings for drug delivery.

SELECTION OF CITATIONS
SEARCH DETAIL
...