Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Neurobiol Aging ; 70: 276-290, 2018 10.
Article in English | MEDLINE | ID: mdl-30055413

ABSTRACT

Alzheimer's disease (AD) is a disease of aging that results in cognitive impairment, dementia, and death. Pathognomonic features of AD are amyloid plaques composed of proteolytic fragments of the amyloid precursor protein (APP) and neurofibrillary tangles composed of hyperphosphorylated tau protein. One type of familial AD occurs when mutant forms of APP are inherited. Both APP and tau are components of the microtubule-based axonal transport system, which prompts the hypothesis that axonal transport is disrupted in AD, and that such disruption impacts cognitive function. Transgenic mice expressing mutated forms of APP provide preclinical experimental systems to study AD. Here, we perform manganese-enhanced magnetic resonance imaging to study transport from hippocampus to forebrain in four cohorts of living mice: young and old wild-type and transgenic mice expressing a mutant APP with both Swedish and Indiana mutations (APPSwInd). We find that transport is decreased in normal aging and further altered in aged APPSwInd plaque-bearing mice. These findings support the hypothesis that transport deficits are a component of AD pathology and thus may contribute to cognitive deficits.


Subject(s)
Aging/physiology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Axonal Transport , Hippocampus/metabolism , Prosencephalon/metabolism , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Hippocampus/pathology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/metabolism , Neural Pathways/pathology , Prosencephalon/pathology
2.
J Immunol ; 196(1): 328-35, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26608923

ABSTRACT

Hyperlipidemia has been extensively studied in the context of atherosclerosis, whereas the potential health consequences of the opposite extreme, hypolipidemia, remain largely uninvestigated. Circulating lipoproteins are essential carriers of insoluble lipid molecules and are increasingly recognized as innate immune effectors. Importantly, severe hypolipidemia, which may occur with trauma or critical illness, is clinically associated with bacterial pneumonia. To test the hypothesis that circulating lipoproteins are essential for optimal host innate defense in the lung, we used lipoprotein-deficient mice and a mouse model of Staphylococcus aureus pneumonia in which invasive infection requires virulence factor expression controlled by the accessory gene regulator (agr) operon. Activation of agr and subsequent virulence factor expression is inhibited by apolipoprotein B, the structural protein of low-density lipoprotein, which binds and sequesters the secreted agr-signaling peptide (AIP). In this article, we report that lipoprotein deficiency impairs early pulmonary innate defense against S. aureus quorum-sensing-dependent pathogenesis. Specifically, apolipoprotein B levels in the lung early postinfection are significantly reduced with lipoprotein deficiency, coinciding with impaired host control of S. aureus agr-signaling and increased agr-dependent morbidity (weight loss) and inflammation. Given that lipoproteins also inhibit LTA- and LPS-mediated inflammation, these results suggest that hypolipidemia may broadly impact posttrauma pneumonia susceptibility to both Gram-positive and -negative pathogens. Together with previous reports demonstrating that hyperlipidemia also impairs lung innate defense, these results suggest that maintenance of normal serum lipoprotein levels is necessary for optimal host innate defense in the lung.


Subject(s)
Bacterial Proteins/metabolism , Hypolipoproteinemias/immunology , Lipoproteins, LDL/blood , Pneumonia, Staphylococcal/immunology , Quorum Sensing/immunology , Staphylococcus aureus/immunology , Trans-Activators/metabolism , Animals , Apolipoproteins B/immunology , Bacterial Proteins/genetics , Cell Line , Disease Models, Animal , Humans , Hypolipoproteinemias/genetics , Immunity, Innate/immunology , Lipoproteins, LDL/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Trans-Activators/genetics
3.
PLoS Pathog ; 9(2): e1003166, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23459693

ABSTRACT

Staphylococcus aureus contains an autoinducing quorum-sensing system encoded within the agr operon that coordinates expression of virulence genes required for invasive infection. Allelic variation within agr has generated four agr specific groups, agr I-IV, each of which secretes a distinct autoinducing peptide pheromone (AIP1-4) that drives agr signaling. Because agr signaling mediates a phenotypic change in this pathogen from an adherent colonizing phenotype to one associated with considerable tissue injury and invasiveness, we postulated that a significant contribution to host defense against tissue damaging and invasive infections could be provided by innate immune mechanisms that antagonize agr signaling. We determined whether two host defense factors that inhibit AIP1-induced agrI signaling, Nox2 and apolipoprotein B (apoB), also contribute to innate control of AIP3-induced agrIII signaling. We hypothesized that apoB and Nox2 would function differently against AIP3, which differs from AIP1 in amino acid sequence and length. Here we show that unlike AIP1, AIP3 is resistant to direct oxidant inactivation by Nox2 characteristic ROS. Rather, the contribution of Nox2 to defense against agrIII signaling is through oxidation of LDL. ApoB in the context of oxLDL, and not LDL, provides optimal host defense against S. aureus agrIII infection by binding the secreted signaling peptide, AIP3, and preventing expression of the agr-driven virulence factors which mediate invasive infection. ApoB within the context of oxLDL also binds AIP 1-4 and oxLDL antagonizes agr signaling by all four agr alleles. Our results suggest that Nox2-mediated oxidation of LDL facilitates a conformational change in apoB to one sufficient for binding and sequestration of all four AIPs, demonstrating the interdependence of apoB and Nox2 in host defense against agr signaling. These data reveal a novel role for oxLDL in host defense against S. aureus quorum-sensing signaling.


Subject(s)
Apolipoproteins B/metabolism , Bacterial Proteins/metabolism , Membrane Glycoproteins/physiology , NADPH Oxidases/physiology , Quorum Sensing/physiology , Receptors, LDL/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Trans-Activators/metabolism , Animals , Blotting, Western , Disease Models, Animal , Female , Gene Expression Regulation, Bacterial , Immunity, Innate , Immunoassay , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Staphylococcal Infections/metabolism , Staphylococcal Infections/pathology , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL