• No results found

Protective properties of lysozyme on β-amyloid pathology : implications for Alzheimer disease

N/A
N/A
Protected

Academic year: 2021

Share "Protective properties of lysozyme on β-amyloid pathology : implications for Alzheimer disease"

Copied!
13
0
0

Loading.... (view fulltext now)

Full text

(1)

Protective properties of lysozyme on β-amyloid

pathology: implications for Alzheimer disease

Linda Helmfors, Andrea Boman, Livia Civitelli, Sangeeta Nath, Linnea Sandin, Camilla

Janefjord, Heather McCann, Henrik Zetterberg, Kaj Blennow, Glenda Halliday, Ann-Christin

Brorsson and Katarina Kågedal

Linköping University Post Print

N.B.: When citing this work, cite the original article.

Original Publication:

Linda Helmfors, Andrea Boman, Livia Civitelli, Sangeeta Nath, Linnea Sandin, Camilla

Janefjord, Heather McCann, Henrik Zetterberg, Kaj Blennow, Glenda Halliday, Ann-Christin

Brorsson and Katarina Kågedal, Protective properties of lysozyme on β-amyloid pathology:

implications for Alzheimer disease, 2015, Neurobiology of Disease, (83), 122-133.

http://dx.doi.org/10.1016/j.nbd.2015.08.024

Copyright: 2015 The Authors. Published by Elsevier Inc. This is an open access article under

the CC BY license.

http://www.elsevier.com/

Postprint available at: Linköping University Electronic Press

http://urn.kb.se/resolve?urn=urn:nbn:se:liu:diva-122341

(2)

Protective properties of lysozyme on

β-amyloid pathology: implications

for Alzheimer disease

Linda Helmfors

a,1

, Andrea Boman

b,1

, Livia Civitelli

b

, Sangeeta Nath

b

, Linnea Sandin

b

, Camilla Janefjord

b

,

Heather McCann

c

, Henrik Zetterberg

d,e

, Kaj Blennow

d

, Glenda Halliday

c

,

Ann-Christin Brorsson

a,

,1

, Katarina Kågedal

b,

⁎⁎

,1

aDivision of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden

bExperimental Pathology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, 581 85 Linköping, Sweden c

Neuroscience Research Australia and University of New South Wales, Randwick New South Wales 2031, Australia

d

Clinical Neurochemistry Laboratory, Department of Neuroscience and Physiology, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden

e

UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom

a b s t r a c t

a r t i c l e i n f o

Article history: Received 14 June 2015 Revised 3 August 2015 Accepted 21 August 2015 Available online 1 September 2015 Keywords: Lysozyme Biomarker Alzheimer disease Drosophila Aβ aggregation

The hallmarks of Alzheimer disease are amyloid-β plaques and neurofibrillary tangles accompanied by signs of neuroinflammation. Lysozyme is a major player in the innate immune system and has recently been shown to prevent the aggregation of amyloid-β1-40in vitro. In this study we found that patients with Alzheimer disease

have increased lysozyme levels in the cerebrospinalfluid and lysozyme co-localized with amyloid-β in plaques. In Drosophila neuronal co-expression of lysozyme and amyloid-β1-42reduced the formation of soluble and

insol-uble amyloid-β species, prolonged survival and improved the activity of amyloid-β1-42transgenicflies. This

suggests that lysozyme levels rise in Alzheimer disease as a compensatory response to amyloid-β increases and aggregation. In support of this, in vitro aggregation assays revealed that lysozyme associates with amyloid-β1-42and alters its aggregation pathway to counteract the formation of toxic amyloid-β species. Overall, these

studies establish a protective role for lysozyme against amyloid-β associated toxicities and identify increased ly-sozyme in patients with Alzheimer disease. Therefore, lyly-sozyme has potential as a new biomarker as well as a therapeutic target for Alzheimer disease.

© 2015 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

1. Introduction

Alzheimer disease is the primary cause of dementia and is manifest-ed as an accumulation of oligomers and extracellular plaques composmanifest-ed of amyloid-β (Aβ), intraneuronal neurofibrillary tangles composed of hyperphosphorylated tau, synaptic failure and progressive neurodegen-eration (Blennow et al., 2006). There are several risk factors proposed for sporadic Alzheimer disease, with ageing and neuroinflammation suggested to be important contributors to the disease (Akiyama et al., 2000; Heneka and O'Banion, 2007; McGeer and McGeer, 2007). There is evidence that Aβ deposits in the brain trigger neuroinflammation via activation of the microglia and astrocytes that surround amyloid

plaques (Halle et al., 2008; Wyss-Coray and Rogers, 2012). However, whether inflammation is the driving force, a contributor or a secondary effect of Alzheimer disease pathology is not known. Sequential cleavage of the amyloid precursor protein (APP) generates Aβ; and the accumu-lation of Aβ is central to Alzheimer disease pathogenesis (Hardy and Selkoe, 2002). Several Aβ isoforms are observed, with Aβ1-40, Aβ1-42,

Aβ1-43and the N-terminally truncated isoforms Aβ3-42and Aβ11-42

being neurotoxic (Jonson et al., 2015) and Aβ1-42is the most abundant

isoform during Alzheimer disease and is highly aggregation prone. Dur-ing Aβ aggregation, monomers turn into oligomers, prefibrillar species, fibrils and insoluble plaques. Aβ aggregation is characterized by a lag phase in which seeds are formed and a logarithmic phase in which the seeds elongate intofibrils that catalyze further fibrillization, which in turn accelerates the aggregation process (Wogulis et al., 2005). In the Alzheimer disease brain, Aβ1-42is the main constituent of plaques

(Portelius et al., 2010). Intermediate structures of aggregated Aβ species generated prior to the formation of the insoluble plaques; oligomers are cytotoxic and cause phosphorylation of tau, synaptic failure and neuro-nal death (Lesne et al., 2013). Several endogenous proteins can manip-ulate the aggregation process of Aβ. The physical interaction between

⁎ Correspondence to: A.-C. Brorsson, Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.

⁎⁎ Correspondence to: K. Kågedal, Division of Cell biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.

E-mail addresses:ann-christin.brorsson@liu.se(A.-C. Brorsson),

katarina.kagedal@liu.se(K. Kågedal).

1

These authors contributed equally to this study.

Available online on ScienceDirect (www.sciencedirect.com).

http://dx.doi.org/10.1016/j.nbd.2015.08.024

0969-9961/© 2015 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Contents lists available atScienceDirect

Neurobiology of Disease

(3)

Aβ and the chaperones clusterin, αB-crystalline, haptoglobin, and α2

-macroglobulin can inhibit Aβ aggregation; thus, these chaperones may play an important role in Alzheimer disease pathogenesis (Matsubara et al., 1996; Du et al., 1998; Barral et al., 2004; Raman et al., 2005; Yerbury et al., 2009).

Lysozyme is part of the innate immune system and possesses bacte-riolytic properties capable of hydrolyzing peptidoglycans in the bacteria cell wall (Fleming, 1922). Lysozyme is secreted by epithelial cells, mac-rophages, astrocytes and microglia, and the enzyme is abundant in various tissues andfluids including liver, spleen, milk, tears, saliva and CSF (Ganz, 2004). Lysozyme retains both oxidant and anti-inflammatory properties (Ogundele, 1998; Liu et al., 2006; Lee et al., 2009), and the level of lysozyme has been reported to be increased in CSF during inflammation (Hällgren and Venge, 1982). Lysozyme was re-cently shown to inhibit Aβ1-40aggregation via binding to the

monomer-ic form of Aβ1-40(Luo et al., 2013; Luo et al., 2014) and has also been

predicted to prevent the aggregation of Aβ17-42via binding to

mono-meric Aβ species (Das et al., 2014). Moreover, in a recent large microar-ray investigation of more than 12,500 genes infive Alzheimer mouse models lysozyme together with e.g. TREM2 were some of the key im-mune genes identified to be overexpressed (Matarin et al., 2015).

In this study we investigated whether lysozyme is activated in Alzheimer disease. Using Alzheimer disease patient samples, we discov-ered that the level of lysozyme was significantly increased in the CSF and that lysozyme co-localized with Aβ1-42in plaques. In a Drosophila

model of Aβ1-42toxicity, lysozyme co-expression reduced Aβ1-42levels,

extended survival and improved the activity of theflies. Using aggrega-tion assays we revealed that lysozyme binds with Aβ1-42inhibiting its

aggregation, and in neuronal cultures this has a neuroprotective role. Our results provide evidence that lysozyme is activated in Alzheimer disease, and therefore has potential as a biomarker. Its role appears to be neuroprotective for increasing Aβ toxicity identifying a potential new therapeutic pathway for the disease.

2. Materials and methods

2.1. Human study populations

2.1.1. CSF cohorts and lysozyme measurements

This study was performed on two separate sets of de-identified, archived matched CSF and serum samples. The study was approved by the Ethical committee at the University of Gothenburg. All samples were provided by the Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital/Mölndal, Sweden. The samples were collected from patients who sought medical advice because of minor cognitive or neurological symptoms. Following routine clinical assessment, the CSF biomarkers for Alzheimer's disease; P-tau181P, T-tau and Aβ1–42

(Hansson et al., 2006) were analyzed in these patients, and based on their neurochemical profile, the samples were divided into three groups; those with CSF biomarkers in the Alzheimer's disease range (AD, n = 10), those with CSF biomarkers in the control range (C, n = 10) and those with CSF biomarkers indicative of neuronal injury but not Alzheimer's disease (having high T-tau, n = 5). The samples with Alzheimer's disease were designated according to CSF biomarker levels using cutoffs that have 95% sensitivity and 80% specificity, includ-ing T-tau 350 ng/l, Aβ1–42530 ng/l and P-tau181P60 ng/l (Hansson et al.,

2006). For further description of patient information, CSF collection and protein analysis via ELISA and Western blotting, see the previously de-scribed methods (Armstrong et al., 2014). The primary antibodies used for Western blotting were lysozyme (A0099, Dako, Glostrup, Denmark) and GAPDH (247 002, Synaptic Systems GmbH, Göttingen, Germany). Equal sample loading was verified by Ponceau S (Sigma-Al-drich, St. Louis, MO, USA) staining of total protein in each lane on the membranes. Thefilms were scanned and the immunoblots were quan-tified using the Image J program (available athttp://rsbweb.nih.gov/ij/). The relative amount of protein corresponding to an immunoreactive

band was calculated as a product of average optical density of the area of the band.

2.1.2. Autopsy confirmed cohorts with brain samples and tissue localization methods

Tissues were received from the Sydney Brain Bank at Neuroscience Research Australia and the New South Wales Tissue Resource Centre at the University of Sydney which are supported by the National Health and Medical Research Council of Australia (NHMRC), University of New South Wales, Neuroscience Research Australia, Schizophrenia Research Institute and National Institute of Alcohol Abuse and Alcoholism (NIH (NIAAA) R24AA012725). Brain tissue collection procedures were ap-proved by the University of New South Wales Human Research Ethics Committee and informed consent was obtained from all individuals prior to donation of their brain. Cases were selected based on their char-acterization with Alzheimer disease according to established neuro-pathological criteria (Montine et al., 2012).

For immunohistochemistry, formalin-fixed, paraffin-embedded 10μm sections of inferior temporal cortex tissues underwent heat-induced antigen retrieval with citrate buffer (pH 6.0) for 3 min, followed by formic acid pretreatment for 3 min. Then, sections were sequentially double-labeled with rabbit monoclonal anti-human lysozyme (1:100, Abcam, Cambridge, UK) visualized with the NovoLink Polymer Detec-tion System (RE7150-K, Leica Biosystems, Wetzlar, Germany). Double labeling was performed with the lysosome antibody and mouse mono-clonal anti-human Aβ1–42(1:200, M0872, Dako, Glostrup, Denmark)

vi-sualized with donkey anti-rabbit Alexa Fluor 568 (1:250, A-10,042, Molecular Probes, Waltham, MA, USA) and goat anti-mouse Alexa Fluor® 488 (1:500, A-11,001, Molecular Probes, Waltham, MA, USA) respectively. Sections were coverslipped with VectaShield for fluores-cence (H-1000, Vector, Burlingame, CA, USA) and examined on a confo-calfluorescence microscope (Nikon C1si). Control experiments for the double labeling were performed with only a single or no primary anti-body with the result of single or no protein labeling observed.

2.1.3. Lysozyme detection by Mesoscale Discovery (MSD)

The MSD technique is similar to the ELISA technique but with electrochemiluminescent detection of the analytes. A standard binding 96-well multi-array plate (L15XA-6, MSD) was coated with 25μl of 15μg/ml cAbHuL6 (the N-terminal domain of a camelid heavy-chain antibody specific for human lysozyme (Dumoulin et al., 2003) (1 h, RT), then the plate was washed with 150μl PBS-T. A total of 25 μl block solution (1% milk in PBS-T) was added to the wells and incubated (1 h, RT), then 20μl CSF was added to the plate and incubated (1 h, RT). The plate was washed and 25μl rabbit anti-human lysozyme antiserum (1:1000 in 1% milk in PBS-T) was added to the wells and incubated (1 h, RT). The plate was washed, and 25μl Sulfo-Tag goat anti-rabbit antibody (R32AB-5, MSD, 1:500 in 1% milk in PBS-T) was added and the plate was incubated (1 h, RT). The plate was washed, and 150μl 2X read buffer (R92TC-2, MSD) was added. The plate was analyzed using a SECTOR Im-ager 2400 instrument (MSD).

Tenfly heads of each genotype were snap-frozen, 150 μl PBS-PI (PBS buffer with Complete EDTA-free Protease Inhibitor Cocktail Tablets, Roche Diagnostics, Basel, Switzerland) was added and thefly heads were homogenized. The samples were centrifuged (13,000 rpm, 1 min); the supernatant constituted the soluble fraction. Lysozyme levels were detected as described above.

2.1.4. Cell culture

Human SH-SY5Y neuroblastoma cells were cultured in Dulbecco's Modified Eagle Medium:Nutrient Mixture F-12 GlutaMAX™ supple-mented with 100 U/ml penicillin, 100μg/ml streptomycin, 1% MEM non-essential amino acids solution and 10% fetal bovine serum in a humidified atmosphere with 5% CO2. To achieve a neuronal-like

pheno-type, the cells were pre-treated with 10μM retinoic acid (Sigma-Al-drich, St. Louis, MO, USA) for 7 days prior to the experiments. Cell

(4)

viability was determined by the XTT assay (Roche) according to the manufacturer's instructions. The XTT absorbance was measured at 450–750 nm using a Victor Wallac plate reader (PerkinElmer, Waltham, MA, USA). Human neuroglioma H4APPswecells were cultured in

Opti-MeMRsupplemented with 100 U/ml penicillin, 100μg/ml streptomycin,

2.5μg/ml blasticidin, 200 μg/ml hygromycin B and 10% fetal bovine serum in a humidified atmosphere with 5% CO2.

2.1.5. Oligomeric Aβ1-42preparation

Freshly made oligomeric Aβ1–42was prepared for each experiment.

Recombinant Aβ1–42(rPeptide) was lyophilized in hexafluoroisopropanol

(HFIP) and dissolved in DMSO to 1.5 mM. Aβ1-42was vortexed and

dilut-ed with cold HEPES buffer to a concentration of 100μM, vortexed 30 s, then sonicated 2 min and incubated 24 h at 4 °C. Size exclusion chroma-tography and electron microscopy confirmed the formation of oligomers (Domert et al., 2014).

2.1.6. Fluorescent labeling of lysozyme

The free amine of the lysine on lysozyme was labeled using the Alexa Fluor® 488 protein labeling kit (Life Technologies). Sodium bicarbonate buffer (pH 8.5) was used to prevent aggregation during the labeling process. The labeled proteins were separated from the free dye by gel chromatography using a PD-10 column eluted with HEPES buffer at pH 7.4. The concentration of Alexa-488-labeled lysozyme was mea-sured on a Nanodrop ND-1000 spectrophotometer (Saveen Werner, Limhamn, Sweden) and calculated by subtracting the contribution of Alexa-488 at 280 nm from the observed absorbance using the following equation:

A280 lysozymeð Þ¼ A280 observedð Þ−A495 Alexa 488ð Þϵ 280 Alexa 488ϵ 495 Alexa 488ðð ÞÞ

The concentration was calculated using the extinction coefficient of ly-sozyme as 2.64 ml mg−1cm−1at 280 nm and the 71,000 cm−1M−1M extinction coefficient of the Alexa 488 fluor probe. The calculated labeling percentage was ~58%.

2.1.7. Fluorescence resonance energy transfer (FRET) using steady state fluorescence spectroscopy

The intermolecular interaction between lysozyme and Aβ species was studied by FRET using steady-statefluorescence spectroscopy. Fluores-cence emission spectra measurements were performed in a Tecan Safire2 multiplate reader (Tecan Group Ltd., Männedorf, Switzerland). Alexa-488 (Exmax494 nm and Emmax520 nm)-labeled lysozyme was used as

the donor and N-terminal TAMRA (carboxytetramethylrhodamine)-labeled synthetic Aβ1-42(Innovagen, Lund, Sweden) was used as the

ac-ceptor (Exmax555 nm and Emmax580 nm) (Biskup et al., 2007, Koch

et al., 2008). Before the measurements, lysozyme-Alexa-488 (200 nM) was incubated for 1 h at 37 °C with 8μM of monomeric Aβ-TAMRA in HEPES buffer (pH 7.4). Size exclusion chromatography confirmed that more than 99% of the Aβ-TAMRA was monomers using a Superdex 75 10/300 GL column (GE Healthcare) equilibrated in PBS.

2.1.8. ThT aggregation assay

Recombinant Aβ1-42(rPeptide) lyophilized from HFIP was dissolved

in 2 mM NaOH to a concentration of 222μM. Aβ1-42was diluted to a

final concentration of 10 μM and then aggregated alone or with 10 or 40μM lysozyme (Sigma-Aldrich) in the presence of 0.3 μM ThT. The samples were loaded in a Corning 96-well black-well clear-bottom mi-crotiter plate (Corning Inc. Life Sciences, Tewksbury, MA, USA), and the in situ change in ThTfluorescence was recorded under quiescent condi-tions at 37 °C for 24 h usingfluorescence spectroscopy (Tecan). The ex-citation was performed at 440 nm and the emission intensity were recorded at 480 nm. For the ThT curves of Aβ aggregated alone or in the presence of lysozyme (10μM), the rate of aggregation (k) was ex-tracted from the maximal slope of the sigmoidal curve and the length of the lag time (tl) was determined byfitting the initial data to a straight

line and a tangent to the steepest region of the growth curve; tlis

de-fined as the time point where the straight line and the tangent intersect. 2.1.9. Transmission electron microscopy (TEM) of end products from the ThT aggregation assay

Samples collected from the 24 h ThT aggregation assay were snap-frozen in liquid nitrogen. A total of 10μl of each sample was added to formvar/carbon-coated 400 mesh copper transmission electron micro-scope grids (Agar Scientific Ltd., Stansted, UK) for 2 min. The fluid was removed and 10μl of 4% uranyl acetate was added for 2 min. The grids were analyzed with a JEOL JEM1230 transmission electron microscope (JEOL Ltd., Tokyo, Japan) equipped with a SC1000 ORIUS CCD camera and the DigitalMicrograph (DM) v.1.71.38 software (Gatan Inc., Pleas-anton, CA, USA).

2.1.10. Immunoanalyses of end products from the ThT aggregation assay A total of 20μl of the 24 h product formed in the ThT aggregation assay was transferred for each reaction. The samples were incubated for 1 h at RT with an antibody solution containing mouse monoclonal anti-Aβ antibody (1:500 in PBS; Mabtech, Nacka strand, Sweden) and rabbit monoclonal anti-lysozyme antibody (1:500 in PBS; Abcam). The samples were centrifuged for 10 min at 13,000 rpm and the supernatant was discarded. The pellet was resuspended in a secondary antibody so-lution containingfluorescently labeled goat anti-mouse (1:600 in PBS; Alexa Fluor 594 goat anti-mouse; Life technologies) andfluorescently labeled goat rabbit (1:600 in PBS; Alexa Fluor 488 goat anti-rabbit; Life technologies) and incubated for 1 h at RT. The samples were centrifuged for 10 min at 13,000 rpm and the supernatant was discarded. A total of 5μl of the pellet was transferred onto SuperFrost Plus slides (Menzel-Gläser, Braunschweig, Germany) and allowed to dry. The slides were mounted usingfluorescence mounting medium (Dako) and analyzed using a Zeiss LSM 780 confocal microscope (Zeiss, Oberkochen, Germany).

The ELISA analyses of the 24 h end point samples were performed by a previously described method that specifically detects the aggregated forms of Aβ without detecting monomers (Höltta et al. 2013). The same monoclonal anti-Aβ N-terminal specific 82E1 antibody (IBL Inter-national, Hamburg, Germany) was used both for capture and detection.

2.1.11. Fly stocks

The Gal4/UAS system was used for CNS-specific expression of UAS transgenes in Drosophila melanogaster (Brand and Perrimon, 1993). The C155-gal4 driver (Lin and Goodman, 1994) was used to express the transgenes in the CNS. Strains of C155-gal4flies carrying UAS-containing genes encoding human wild type lysozyme (Kumita et al. 2012), Aβ1-42(Crowther et al., 2005) and both lysozyme and Aβ1-42

(both containing a secretion-tag) were generated. Background C155-gal4 w1118was used to generate controlflies. D. melanogaster stocks were allowed to develop under a 12:12 h light:dark cycle at 29 °C at 60% humidity. Theflies were kept in 50 ml plastic vials containing stan-dard Drosophila food (corn meal, molasses, yeast and agar) and main-tained post-eclosion in 50 ml plastic vials containing six ml agar (20 g agar and 20 g sugar dissolved in 1 l water) and yeast paste (dry Baker's yeast dissolved in water) at 29 °C under 12:12 h light:dark cycle.

2.1.12. Drosophila longevity and locomotor assays

One hundred Drosophilaflies of each genotype were divided into plastic vials containing agar food and yeast paste with tenflies in each vial and kept at 29 °C in a 12:12 h light:dark cycle. Every 2–3 days the flies were transferred to fresh food and the numbers of flies alive were counted. Prism GraphPad software 6 (GraphPad Software, San Diego, CA USA) was utilized to graph Kaplan-Meier survival curves (Kaplan and Meier, 1958) and to run the log rank statistical analysis.

To analyze the locomotor behavior of theflies, a locomotor assay using the iFly software (Jahn et al., 2011), was used. For each genotype, three vials with tenflies in each were collected and filmed for 90 s. The

(5)

flies were tapped to the bottom every 30 s (re-activating the locomotor behavior), yielding three video clips with three clips of 30 s each. The videos were processed and analyzed using the iFly software, which cal-culated the velocities and the angle of movement for each clip. The data were plotted using GraphPad Prism 6.

2.1.13. Immunohistochemistry offly heads

Drosophila heads were embedded on the day of eclosion or at day 20 in Tissue-Tek OCT compound (Histolab, Gothenburg, Sweden) using Cryomold specimen molds and stored at−80 °C until use. The OCT blocks were sectioned using a Microm HM 550 Cryostat (Microm Inter-national, Walldorf, Germany) into 20μm thin sections that were placed on SuperFrost Plus slides (Menzel-Gläser) and stored at−20 °C until use. The sections werefixed in 4% w/v PFA in PBS for 10 min at RT. The slides were washed in PBS (3 × 3 min) followed by a (1 × 3 min) wash in PBS-T (PBS with 0.05% v/v Tween-20) before blocking in 10% w/v BSA in PBS-T for 1 h at RT. Mouse monoclonal anti-Aβ antibody (1:500 in 1% BSA w/v in PBS-T; Mabtech) and rabbit monoclonal anti-lysozyme (1:500 in 1% BSA w/v in PBS-T; Abcam) were added to the slides and incubated for 1 h at RT. The slides were washed with PBS-T (3 × 5 min). Goat anti-mousefluorescently labeled secondary antibody (1:600 in 1% w/v BSA in PBS-T; Alexa Fluor 594 goat anti-mouse; Life Technologies) and goat anti-rabbitfluorescently labeled secondary anti-body (1:600 in 1% w/v BSA in PBS-T; Alexa Fluor 488 goat anti-rabbit; Life Technologies) were applied to the slides and incubated for 1 h at RT. The slides were mounted using Vectashield with DAPI (Vector Lab-oratories). The slides were analyzed using a Zeiss LSM 780 confocal mi-croscope (Zeiss). The micrographs were processed using Adobe Photoshop (Adobe Systems, San Jose, CA, USA); all images were treated identically.

2.1.14. Analyses of Aβ1-42levels in Drosophila

Fivefly heads for each genotype were placed in Eppendorf tubes and snap frozen, and then analyzed on MSD as described previously (Caesar et al., 2012). To adjust for variation in the protein extraction step, a quantitation of the total amount of protein from each sample offly ho-mogenate was performed using the Bio-Rad DC Protein Assay Kit II (500–0112, Bio-Rad, Hercules, CA, USA).

2.1.15. Lysozyme expression in cells and binding tofibrillar Aβ

Neuroblastoma and neuroglioma cells were incubated with oligo-meric Aβ1–42diluted in serum-free culture media at afinal

concentra-tion of 1μM for 1 h. After extensive PBS washing and trypsinization, the cells were re-seeded and grown in serum-free media for 24 or

48 h. The cells were lysed, and lysozyme levels were measured via Western blotting as described previously (Armstrong et al., 2014).

A total of 50μg recombinant Aβ1-42-TAMRA lyophilized from HFIP

was dissolved in 2.2μL DMSO, diluted to 100 μM in 20 mM HEPES (pH 7.4), vortexed, and aggregated at 37 °C for 24 h. The aggregated fi-brillar Aβ1-42-TAMRA was diluted to afinal concentration of 10 μM and

incubated 1 h at RT alone or with 40μM lysozyme-Alexa 488 (Sigma-Al-drich). The samples were centrifuged for 10 min at 13,000 rpm and the supernatant was discarded. A total of 5μl of the pellet was transferred onto SuperFrost Plus slides (Menzel-Gläser) and allowed to dry. The slides were mounted usingfluorescence mounting medium (Dako) and sealed with nail Polish and were analyzed using a Zeiss LSM 510 META confocal microscope (Zeiss).

2.2. Statistical analysis

All statistical analyses were performed using GraphPad Prism Software.

The mean value and standard deviation or standard error of the mean were calculated for all data. The nonparametric Mann–Whitney U test was used to test for significant differences between groups. For Drosophila longevity statistics, Kaplan–Meier survival curves and log-rank (Mantel–Cox) statistical analyses were performed. Statistical sig-nificance was defined for p-values less than or equal to 0.05 (*), 0.01 (**) and 0.001 (***).

3. Results

3.1. Lysozyme is increased in CSF and present in plaques from patients with Alzheimer disease

Lysozyme levels were tested in two cohorts of CSF samples collected from patients with Alzheimer disease and controls (Table 1, CSF set 1 and 2). Lysozyme levels were significantly higher in the CSF samples from Alzheimer patients (Fig. 1A and B). This was demonstrated using two different methods: Western blotting with quantification as shown inFig. 1A and the Meso Scale Discovery technique for the second valida-tion cohort of CSF samples as shown inFig. 1B. Lysozyme levels did not differ between the control and Alzheimer disease cohorts in matched serum samples (Fig. 1B). To investigate whether lysozyme up-regulation was due to general neurodegeneration, lysozyme levels were analyzed in the CSF of a third set of non- Alzheimer disease pa-tients who had normal Aβ1-42and P-tau181Plevels but elevated total

tau (T-tau). No significant difference between lysozyme levels were

Table 1

Study demographics of Alzheimer disease (AD) patients versus control (C) individuals CSF set 1, CSF set 2, and CSF set 3 (C 1-5 from set 1 versus High T-tau patients). Data are presented as the mean and (range).

Study group Age years Sex

M:F

CSF T-tau ng/l CSF Aβ42 ng/l CSF P-tau181Png/l CSF/serum

Albumin ratio CSF set 1 AD n = 10 68.5 (58–89) 5:5 880 (580–1680) 330 (310–390) 123 (84–177) 5.5 (4.2–9.5) C n = 10 74 (43–82) 6:4 295 (160–390) 780 (553–1140) 42 (26–61) 7.8 (3.8–10.8) CSF set 2 AD n = 10 73 (50–89) 4:6 844 (467–1350) 342 (200–440) 97 (76–165) 7.6 (6.1–10.0) C n = 10 64 (50–76) 6:4 245 (141–363) 821 (564–1150) 39 (23–60) 5.7 (4.0–7.1) CSF set 3 C 1-5 set 1 n = 5 72 (61–79) 3:2 271 (170–380) 705 (553–970) 42 (26–57) 6.7 (3.8–9.2) High-T-tau n = 5 65.2 (26–82) 2:3 2541 (487–9620) 466 (334–576) 73 (62–80) No data available

(6)

detected between the control and high-T-tau samples (Fig. 1C), demon-strating that the increase in lysozyme in Alzheimer disease CSF was not caused by generalized neuronal damage.

To study whether lysozyme and Aβ1-42 co-localize in plaques

in Alzheimer disease, immunohistochemistry was performed using postmortem formalin-fixed and paraffin-embedded inferior temporal

Fig. 2. Co-localization of lysozyme and Aβ1-42in Alzheimer disease plaques. A) Representative image of peroxidase immunohistochemistry with an antibody specific for lysozyme, showing

abundant staining of neuritic (arrowhead) and diffuse (asterisk) plaques in human Alzheimer disease inferior temporal cortex tissue. B) Immunohistochemistry of lysozyme showing the presence of lysozyme around a plaque core (arrowheads). C–E & F–H) Double imunoflourescence labeling of the Aβ1-42 (green) and lysozyme (red) demonstrating co-localization of the two proteins (yellow in merge) in the typical cortical plaques in Alzheimer's disease.

Fig. 1. Lysozyme is up-regulated in CSF from Alzheimer disease patients. A) Lysozyme levels in CSF from ten controls (C) and ten Alzheimer (AD) patients were analyzed using Western blotting followed by densitometric quantification. The blots were cropped. The bars represent the mean ± SD (**p b 0.01). B) Lysozyme levels in CSF and serum from ten controls (C) and ten Alzheimer (AD) disease patients were analyzed using Meso Scale Discovery. The bars represent the mean ± SEM (**pb 0.01). C) The lysozyme levels in five control (C) samples and five samples with neuronal injury but not Alzheimer's disease (High T-tau) were analyzed using Western blotting (the samples were applied with increasing levels of T-tau as illustrated by the triangles) followed by densitometric quantification. The bars represent the mean ± SD.

(7)

cortex tissues from patients with sporadic Alzheimer disease. The stain-ing pattern of lysozyme resembled the morphology of diffuse and neu-ritic plaques (Fig. 2A and B). Double-labeling with antibodies against lysozyme and Aβ1–42revealed that lysozyme was localized with Aβ 1-42in diffuse and neuritic plaques, with clumping of lysozyme around

the plaque core (Fig. 2C–H).

In the CSF from patients with Alzheimer's disease there was no cor-relation between the levels of Aβ1-42and lysozyme, largely due to the

similar concentrations of Aβ1-42(310–390 ng/l) in the CSF samples at

this stage of the Alzheimer's disease. In order to explore whether Aβ could up-regulate the expression and secretion of lysozyme in cell models, both neuroglioma cells and differentiated human neuroblasto-ma cells were exposed for 24 h or 24 and 48 h, respectively, to a subtoxic concentration of 1μM oligomeric Aβ1-42. A significant increase in both

intracellular lysozyme and secreted lysozyme was detected from the neuroglioma cells (Fig. 3A–B). In the neuroblastoma cells, secreted lyso-zyme was not detected in the medium (data not shown) and the

intracellular level of lysozyme was not changed after 24 h, but was significantly increased after 48 h of Aβ1-42exposure (Fig. 3C). This

result demonstrates that Aβ can trigger the expression of lysozyme in cell-based systems and indicates that the increased levels of ly-sozyme in CSF from Alzheimer disease patients might originate from gliacells.

3.2. Lysozyme suppresses the toxicity of Aβ in D. melanogaster

On the basis of these clinical results, we developed a novel lysozyme/ Aβ1-42Drosophila line by co-expressing human lysozyme with Aβ1-42

(lysozyme:Aβ) in the CNS using the neuronal-specific C155-gal4 driver. Both lysozyme and Aβ1-42were expressed with a secretion tag to allow

secretion into the extracellular space. A longevity assay was performed to monitor the toxic effects of the Aβ peptide on fly neurons (Crowther et al., 2005; Hirth, 2010) and to investigate the impact of lysozyme on in vivo Aβ toxicity. The median survival time (represented by the

Fig. 3. Lysozyme is up-regulated in neuroglioma and neuroblastoma cells treated with oligomeric Aβ. Intracellular and secreted levels of lysozyme in cells treated with 1 μM oligomeric Aβ for 24 or 48 h were analyzed using Western blot. Densitometric quantifications were performed on the scanned Western blots and normalized to GAPDH levels or Ponceau S staining. The bars represent the mean ± SD, n = 3 (*pb 0.05). Blots were cropped. A) Lysozyme levels in neuroglioma H4APPswe cells, B) H4APPswe supernatants and C) differentiated SH-SY5Y neu-roblastoma cells.

(8)

death of 50% of theflies) was 28 days for the Aβ flies and 32 days for both the control and lysozyme:Aβ flies (Fig. 4A). Thus, co-expressing ly-sozyme with Aβ resulted in an increased life span of the Aβ flies that equaled the median survival time of the controlflies. To achieve a more complete picture offly health, a locomotor assay was performed using the iFly technique (Jahn et al., 2011) to analyze the velocity and angle of movement. The average velocity for a healthy youngfly is 10 mm/s; as thefly ages, the velocity decreases. Shortly before the flies die they become immobile and their velocity cannot be recorded; thus, a cut-off value of 4 mm/s was set as an indication of disability. The Aβ-expressing flies showed a substantial reduction in their velocity, reaching the cut-off value of 4 mm/s at day 20 compared with day 35 for the controlflies (Fig. 4B), when co-expressing lysozyme with Aβ, this effect was postponed by 6 days and occurred at day 26 (Fig. 4B). The angle of movement describes the deviation of theflies' path from verti-cal when they move from the bottom to the top of the vial (Khabirova, 2012). The angle of movement for a healthy youngfly is approximately 55°, and this angle increases as thefly ages. A cut-off value of 80° was set where values above indicate that the mobility functions of theflies (i.e., orientation and movement direction) are impaired. For the Aβ flies, the angle of movement diverted from that of healthy control flies at day 14 when the cut-off value of 80° was reached. However, for the lysozyme:Aβ flies this day was postponed by 7 days, and the cut-off value was not reached until day 21. The controlflies demonstrated a normal angle of movement until day 34 (Fig. 4C). The protective effects of lysozyme in the longevity and locomotor assays of the Aβ1-42

express-ingflies revealed that lysozyme has a pathomechanistic relevant effect on the reduction of Aβ toxicity.

To investigate the location of Aβ and lysozyme in the fly CNS, sec-tions of Drosophila brains expressing lysozyme, Aβ or both were co-stained with antibodies against lysozyme and Aβ1–42. In flies

co-expressing lysozyme and Aβ1-42, the proteins were in close proximity.

Thisfinding strongly suggests an interaction between these two pro-teins in thefly CNS (Fig. 5).

The effects of lysozyme on the levels of Aβ1-42in Drosophila brains

were also analyzed. Soluble and insoluble Aβ1-42levels were measured

in the Aβ flies with and without co-expression of lysozyme at the day of eclosion (day 0) and 20 days later. For Aβ1-42expressingflies, the

insol-uble fraction of Aβ1-42was higher than the soluble fraction both at days

0 and 20 (Fig. 6A, B); indeed these insoluble species accumulated over time, and a significantly higher level was detected at day 20 compared with day 0 (Fig. 6B). When co-expressing lysozyme and Aβ1-42, the

ac-cumulation of both soluble and insoluble Aβ1-42in agedflies was

signif-icantly reduced compared withflies expressing Aβ1-42alone (Fig. 6A, B).

A reduced level of lysozyme was detected both at day 0 and day 20 in the lysozyme:Aβ flies compared with the lysozyme alone flies; this dif-ference was highly significant at day 20 (Fig. 6C). These analyses clearly show that lysozyme and Aβ are able to interact in the fly CNS and that the presence of lysozyme results in decreased amounts of accumulated Aβ species in the Aβ1-42flies.

3.3. Lysozyme binds to and inhibits the Aβ1-42fibril process and cell death

Next, we studied the in vitro interaction properties between lyso-zyme and Aβ using FRET steady state fluorescence measurements. The decrease of donorfluorescence at 520 nm from lysozyme labeled with Alexa-488 and the increase of acceptorfluorescence at 580 nm from monomeric Aβ1-42labeled with TAMRA demonstrated FRET between

the donor and acceptor due to binding of lysozyme with monomeric Aβ1-42(Fig. 7A). To investigate the capacity of lysozyme to interact

withfibrillar Aβ, 10 μM of TAMRA-labeled Aβ1-42was aggregated for

24 h prior to the addition of Alexa-488–labeled lysozyme (40 μM) for 1 h. Aβ was detected as fibrillar structures, and lysozyme was found as-sociated with these structures (Fig. 7B).

To investigate whether the binding between lysozyme and Aβ has any effect on the aggregation kinetics of Aβ, the fluorescence signal

from thioflavin T (ThT) (Brorsson et al., 2010) was followed during the aggregation of 10μM Aβ1-42in the absence and presence of

lyso-zyme. The Aβ aggregation kinetics were substantially slowed down at a 1:1 ratio between Aβ1-42and lysozyme; the rate of aggregation (k),

which is a measure of the elongation efficiency of fibril formation, was decreased from 17 min−1to 5 min−1and the lag time (tl), which

mir-rors the rate of nucleation events, was prolonged from 4 h to 6 h (Fig. 8A). The end ThT signal was also greatly reduced from 7800 AU for Aβ aggregated alone, to 4200 AU in the presence of an equal concen-tration of lysozyme and to 450 AU in the presence of a four-fold higher concentration of lysozyme (Fig. 8A). No change in the ThT signal was detected for lysozyme alone at the two concentrations used (Fig. S1), having an average signal between 160–180 AU.

Transmission electron microscopy (TEM) images captured at the 24 h time point of the aggregation experiment confirmed fibril forma-tion by the aggregated Aβ1-42sample (Fig. 8D) whilst nofibrils could

be detected for the lysozyme samples (Fig. S2). Fibrillar species were formed in the presence of a 1:1 ratio of the Aβ1-42and lysozyme

concen-trations; these forms were accompanied by smaller aggregated

non-Fig. 4. Protective effect of lysozyme in a Drosophila Alzheimer disease model. A) Survival trajectories forflies expressing Aβ in the CNS in the absence or in the presence of lysozyme expression compared with controlflies. Kaplan–Meier graph showing percent survival vs. age offlies in days (***p b 0.001). Locomotor assays performed using iFly: B) velocities and C) angle of movement recorded over time for Aβ, lysozyme:Aβ and control flies.

(9)

fibrillar species (Fig. 8D). In the presence of four-fold higher lysozyme, the aggregated samples were dominated by non-fibrillar structures (Fig. 8D).

Next, the possible co-localization of Aβ1-42and lysozyme after 24 h

of aggregation was investigated. When Aβ1-42was aggregated in the

presence of lysozyme, aggregates were detected by both anti-Aβ and anti-lysozyme antibodies, thereby demonstrating the co-localization of Aβ1-42and lysozyme. The product formed from the 1:1 ratio of Aβ 1-42and lysozyme appeared as large yellow aggregates where Aβ and

ly-sozyme co-localized; these aggregates were accompanied by some small spheres of Aβ alone (red) and amorphous species of lysozyme alone (green) (Fig. 8E). The product formed from the 1:4 ratio of Aβ and lysozyme appeared as small yellow spheres with almost complete co-localization (Fig. 8E). Using an ELISA assay that specifically detects the aggregated forms of Aβ without detecting monomers (Höltta et al., 2013), we determined that Aβ1-42samples aggregated for 24 h

demonstrated a high level of aggregated Aβ species, whereas Aβ1-42

ag-gregated in the presence of lysozyme had significantly lower levels of these aggregated Aβ forms (Fig. 8D). Thesefindings support the aggre-gation kinetics results (Fig. 8A, D and E), which suggested that lysozyme inhibits thefibrillization process of Aβ1-42.

To investigate whether lysozyme could alter the toxic properties of Aβ, the viability of neuroblastoma cells was monitored, 3 days after ex-posure to Aβ1-42aggregated with or without lysozyme, using XTT. Aβ 1-42was aggregated with or without lysozyme for 6 and 24 h and was

then added to cells. Aβ1-42was toxic after 6 h of aggregation, causing

cells to shrivel up; however, at 24 h the aggregated Aβ1-42species

showed no toxicity (Fig. 8C). Moreover, lysozyme protected the cells against Aβ1-42toxicity at 6 h. The TEM images inFig. 8B show that

small-er non-fibrillar species were formed when Aβ1-42was aggregated in the

presence of four-fold more lysozyme for 24 h. No cell toxicity was de-tected after 24 h of Aβ aggregation with lysozyme, demonstrating that

Fig. 5. Lysozyme and Aβ are localized in close proximity in the Drosophila brain. Drosophila brains collected at day 20 from Aβ, lysozyme, Aβ:lysozyme and control flies were sectioned and co-stained with anti-Aβ (red) and anti-lysozyme (green) antibodies, followed by counterstaining of the cell nuclei with DAPI (blue). Close proximity between Aβ and lysozyme is shown in yellow for the Aβ:lysozyme flies. Sections are from the superior medial protocerebrum and fan-shaped body (inset figure). Scale bars = 50 μm.

(10)

these structures do not possess any cytotoxic properties (Fig. 8C). These results suggest that the binding of lysozyme to Aβ1-42decreases the

ag-gregation propensity, and thereby bypasses the route of formation of toxic Aβ1-42species.

4. Discussion

Triggering of the innate immune system with the secretion of pro-and anti-inflammatory cytokines is an early event in Alzheimer disease. Lysozyme is an important player in the innate immune system but there has not been any proof of a direct relationship between lysozyme and Alzheimer disease. However, in a recent microarray study from a large screen of more than 12,500 genes, the lysozyme gene was identi-fied to be overexpressed in five mice models of Alzheimer disease (Matarin et al., 2015). Two other reports from Luo et al. demonstrate that lysozyme in vitro binds and prevents the aggregation of Aβ1-40.

The present study further reveals the involvement of lysozyme in Alzheimer disease where lysozyme was detected at significantly higher levels in CSF from Alzheimer disease patients compared with controls and was co-localized with Aβ in plaques. We also demonstrate the physiological relevance of lysozyme using a Drosophila model of Aβ 1-42toxicity, where lysozyme overexpression prolonged survival and

enhanced the activity of the Aβ1-42flies. At a mechanistic level, in vitro

assays revealed that lysozyme bound to Aβ and influenced the structure of aggregated Aβ1-42, thereby reducing the toxicity of Aβ1-42.

Because lysozyme levels were increased in CSF from patients with Alzheimer disease, we investigated the Alzheimer disease specificity of lysozyme using CSF from patients with increased CSF levels of T-tau but normal Aβ1–42and P-tau181Pvalues (i.e., patients suffering axonal

damage most likely not caused by Alzheimer disease). Elevated levels of T-tau is found after acute damage to the brain such as stroke and subarachnoid hemorrhage, and the levels of these biomarkers correlate with the severity of the brain damage (Hesse et al., 2000). The lysozyme levels did not increase with increasing levels of T-tau, which implies that increased lysozyme levels in the Alzheimer disease CSF is not secondary to general neurodegeneration. The activation of neuro-inflammatory pathways in the brain is emphasized as a major risk factor for the development of Alzheimer disease. Several reports from trans-genic mice studies indicated that immune activation might prime the brain for Alzheimer disease pathology, such as Aβ plaques and tau ag-gregation (Qiao et al., 2001; Sheng et al., 2003; Kitazawa et al., 2005). If the increased lysozyme levels in CSF from Alzheimer disease patients are triggered by early immune activation, the detection of lysozyme in CSF might be useful as a biomarker for Alzheimer disease particularly in its early preclinical phase. We also showed that lysozyme is closely associated with cortical amyloid plaques, therefore the physiological relevance of lysozyme was investigated in a Drosophila model of Aβ 1-42toxicity. Aβ1-42expressed in thefly CNS resulted in neurological

im-pairments that manifested as effects on the longevity and locomotor as-says. The health of Aβ-expressing flies was remarkably increased when lysozyme was co-expressed with Aβ; the lifespan was extended to that of normalflies and the locomotor activity was substantially improved. The level of insoluble Aβ was found to be considerably higher in the brains of the Aβ1-42flies compared with the soluble levels of the

Fig. 7. Lysozyme associates with monomeric andfibrillar Aβ. A) Fluorescence intensity of 0.2μM Alexa-488 labeled lysozyme (donor) in the absence (black line) and in the presence (red line) of 8μM TAMRA-labeled monomeric Aβ1-42(acceptor). The decrease of donor

fluorescence and increase of acceptor fluorescence indicates fluorescence resonance ener-gy transfer (FRET) between the donor and acceptor due to the association of lysozyme with monomeric Aβ. B) Representative confocal images of 10 μM Aβ1-42labeled with

TAMRA that was aggregated for 24 h and then incubated for 1 h with 40μM lysozyme la-beled with Alexa Fluor 488. The merged image of Aβ1-42and lysozymefluorescence

dem-onstrates co-localization of the two proteins. Scale bar = 20μm. Fig. 6. Lysozyme counteracts the formation of insoluble Aβ species in Drosophila brains.

Levels of Aβ and lysozyme in Drosophila at days 0 and 20 detected by Meso Scale Discov-ery. A) Amounts of soluble Aβ and B) insoluble Aβ in flies expressing Aβ in the absence or in the presence of lysozyme expression compared with controlflies. C) Amounts of soluble lysozyme inflies expressing lysozyme in the absence and in the presence of Aβ expression compared with controlflies. Bars represent means ± SEM, n = 5 (*p b 0.05; **p b 0.01; ***pb 0.001 and ****p b 0.0001).

(11)

peptide, and the insoluble species of the peptide accumulated over time. Thus, it is likely that these insoluble Aβ species or intermediate species formed on the pathway toward insoluble species are cytotoxic and con-tribute to the reduction in lifespan and locomotor activity observed for the Aβ1-42flies. When Aβ was co-expressed with lysozyme, the level

of insoluble Aβ species was reduced in both young and aged flies, indi-cating that the rescue effects in the lysozyme:Aβ flies are caused by the capacity of lysozyme to counteract the formation of these insoluble Aβ species, thereby reducing the formation of toxic Aβ species.

We revealed that lysozyme bound to both monomeric andfibrillar Aβ1-42; this result is in agreement with recent studies where lysozyme

was demonstrated to bind to monomeric Aβ1-40 (Luo et al., 2013,

2014). Additionally, we detected co-localization between lysozyme and Aβ in aggregates formed both in vivo and in vitro. Our observation that lysozyme reduces the aggregation kinetics of Aβ is likely caused by the binding of lysozyme to monomeric Aβ during the initiation of the nucleation phase. In turn, this binding results in a change in the Aβ aggregation process where fibril formation of Aβ is prevented and the formation of aggregated non-fibrillar species, composed of both ly-sozyme and Aβ, is promoted. Studies on cells showed that during the aggregation process of Aβ, cytotoxic aggregated species were formed at 6 h, whereas in the presence of lysozyme no cytotoxic species were formed. This demonstrates that when lysozyme binds to monomeric Aβ, the pool of free monomeric Aβ peptides that can aggregate and form cytotoxic species is reduced in favor of the formation of non-toxic, non-fibrillar lysozyme-Aβ species. The formation of these species is likely to occur via a pathway that differs from the Aβ aggregation pathway towardsfibril formation. Luo et al. recently showed that lyso-zyme also prevents the toxicity of Aβ1-40(Luo et al., 2014). The in vivo

step at which lysozyme was able to rescue Aβ cytotoxicity in the fly CNS is likely to be in line with the in vitro effect of lysozyme on the Aβ aggregation process. By binding of lysozyme to monomeric Aβ in the

fly brain, less Aβ peptides are able to aggregate into cytotoxic species. Instead, the lysozyme-bound Aβ peptides either participate in the for-mation of other, presumptively non-toxic, aggregates composed of both lysozyme and Aβ or are degraded, because the degradation of lyso-zyme and Aβ seems to be more efficient when the proteins are co-expressed compared with when the proteins are co-expressed individually (Kumita et al., 2012).

Little is known about the implications of lysozyme involvement in Alzheimer disease. One pathological feature of Alzheimer disease is the failure of the lysosomal clearance mechanisms that manifests as a severe buildup of lysosomal-related compartments in dystrophic neurites (Nixon et al., 2005). Ourfinding that lysozyme is increased in CSF from Alzheimer disease patients might represent a cellular compen-sation for decreased lysosomal function in the Alzheimer disease brain (Nixon and Yang, 2011) via the up-regulation of lysosomal proteins such as lysozyme (Gupta et al., 1985). The lysosomal proteins LAMP-1, LAMP-2 and cathepsin D have previously been reported to be increased in CSF from Alzheimer disease patients (Schwagerl et al., 1995; Armstrong et al., 2014). Because neuroinflammation is part of the pa-thology of most lysosomal storage disorders, the up-regulation of lyso-somal proteins might be a general phenomenon in these disorders. A functional loss of the lysosomal system may also affect phagocytosis and recycling in microglial cells, which might in turn lead to microglial activation and increased secretion of lysozyme. Another possibility is that the up-regulation of lysozyme might be directly caused by the ac-cumulation of Aβ in Alzheimer disease patients, because our study dem-onstrated that exposure of neuroglia cells to Aβ caused up-regulated expression and secretion of lysozyme. No secretion of lysozyme was de-tected from neuroblastoma cells, indicating that the origin of lysozyme in CSF and plaques is astrocytic rather than neuronal.

Our results demonstrating the rescue effects of lysozyme on Aβ tox-icity establish a potential protective role for lysozyme in Alzheimer

Fig. 8. Effect of lysozyme on Aβ aggregation and cytotoxicity. A) Aggregation of 10 μM Aβ alone or in the presence of 10 μM or 40 μM lysozyme for 24 h probed by ThT. Representative ThT fluorescence curves are shown, n = 3. B) Detection of aggregated Aβ species by ELISA of Aβ aggregated for 24 h alone or with increasing lysozyme concentrations. C) Viability of differ-entiated SH-SY5Y cells analyzed by XTT after 3 days of exposure to Aβ aggregated at 10 μM for 6 or 24 h alone or in the presence 40 μM lysozyme and then diluted 1:10 in cell culture medium. The bars represent the mean ± SD, n = 4 (*pb 0.05, **p b 0.01). D) TEM images of 10 μM Aβ aggregated alone or in the presence of 10 or 40 μM lysozyme. Images were taken at the end of the aggregation experiment in A. Scale bar = 500 nm. E) Fluorescence from the anti-Aβ antibody (red) and anti-lysozyme antibody (green) in samples analyzed at the end of the aggregation experiments in A. Scale bar = 20μm.

(12)

disease pathology. It has been reported that lysozyme apart from its gly-cosidase activity also has antioxidant properties via binding to advanced glycation end products that generate reactive oxidant species (Liu et al., 2006). An increased expression of lysozyme could also be due to the modification of the glycosylation pattern of proteins in Alzheimer dis-ease. APP is glycosylated at Asn467 (Pahlsson et al., 1992), and the dele-tion of Asn467 affects the intracellular sorting of APP (Yazaki et al., 1996). It was also demonstrated that CSF from Alzheimer disease pa-tients has increased levels of glycosylated short Aβ fragments (Halim et al., 2011). However, understanding whether lysozyme binds more ef-ficiently to glycosylated Aβ species or modifies Aβ generation via changes in glycosylation patterns requires further investigation.

We propose that the increased level of lysozyme in CSF is a protec-tive response that eventually becomes overwhelmed by Alzheimer dis-ease pathology. This is supported by the presence of lysozyme in Aβ1-42

plaques in patients with Alzheimer disease. The existence of lysozyme in mature plaques may be a residual effect of unsuccessful inhibition of oligomer formation, perhaps due to insufficient lysozyme levels or the lack of persistence of sufficient levels to achieve this function. Adding ourfindings that lysozyme has an inhibitory effect on Aβ aggre-gation and cytotoxicity to previous reports that lysozyme has anti-inflammatory and anti-oxidative properties indicates that lysozyme is an interesting therapeutic target for Alzheimer disease. To investigate the feasibility of lysozyme as an intervention for Alzheimer disease, ini-tial mechanistic studies should be performed in an APP mouse model to pinpoint the optimal time for intervention, any side effects from altering lysozyme expression as well as toxicity studies for the method of choice for achieving lysozyme overexpression.

Supplementary data to this article can be found online athttp://dx. doi.org/10.1016/j.nbd.2015.08.024.

Conflict of interest

The authors declare no competingfinancial interests. Acknowledgements

The authors thank Damian Crowther for kindly providing Aβ flies and the iFly equipment and Maria Lindbjer-Andersson for performing the ELISA specific for aggregated Aβ.

We are grateful for the brain tissue samples from the Sydney Brain Bank at Neuroscience Research Australia and the New South Wales Tissue Resource Centre at the University of Sydney which are supported by the National Health and Medical Research Council of Australia (NHMRC), University of New South Wales, Neuroscience Research Australia, Schizophrenia Research Institute and National Institute of Al-cohol Abuse and AlAl-coholism (NIH (NIAAA) R24AA012725). GMH is a Senior Principal Research Fellow of the National Health and Medical Re-search Council of Australia (#1079679).

References

Akiyama, H., Barger, S., Barnum, S., Bradt, B., Bauer, J., Cole, G.M., et al., 2000.Inflammation and Alzheimer's disease. Neurobiol. Aging 21 (3), 383–421.

Armstrong, A., Mattsson, N., Appelqvist, H., Janefjord, C., Sandin, L., Agholme, L., et al., 2014.Lysosomal network proteins as potential novel CSF biomarkers for Alzheimer's disease. Neruomol. Med. 16 (1), 150–160.

Barral, J.M., Broadley, S.A., Schaffar, G., Hartl, F.U., 2004.Roles of molecular chaperones in protein misfolding diseases. Semin. Cell Dev. Biol. 15 (1), 17–29.

Biskup, C., Zimmer, T., Kelbauskas, L., Hoffmann, B., Klocker, N., Becker, W., et al., 2007.

Multi-dimensionalfluorescence lifetime and FRET measurements. Microsc. Res. Tech. 70 (5), 442–451.

Blennow, K., de Leon, M.J., Zetterberg, H., 2006.Alzheimer's disease. Lancet 368 (9533), 387–403.

Brand, A.H., Perrimon, N., 1993.Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development 118 (2), 401–415.

Brorsson, A.C., Kumita, J.R., MacLeod, I., Bolognesi, B., Speretta, E., Luheshi, L.M., et al., 2010.Methods and models in neurodegenerative and systemic protein aggregation diseases. Front. Biosci. 15, 373–396.

Caesar, I., Jonson, M., Nilsson, K.P., Thor, S., Hammarstrom, P., 2012.Curcumin promotes A-betafibrillation and reduces neurotoxicity in transgenic Drosophila. PLoS One 7 (2), e31424.

Crowther, D.C., Kinghorn, K.J., Miranda, E., Page, R., Curry, J.A., Duthie, F.A., et al., 2005.

Intraneuronal Abeta, non-amyloid aggregates and neurodegeneration in a Drosophila model of Alzheimer's disease. Neuroscience 132 (1), 123–135.

Das, P., Kang, S.G., Temple, S., Belfort, G., 2014.Interaction of amyloid inhibitor proteins with amyloid beta peptides: insight from molecular dynamics simulations. PLoS One 9 (11), e113041.

Domert, J., Rao, S.B., Agholme, L., Brorsson, A.C., Marcusson, J., Hallbeck, M., et al., 2014.

Spreading of amyloid-beta peptides via neuritic cell-to-cell transfer is dependent on insufficient cellular clearance. Neurobiol. Dis. 65, 82–92.

Du, Y., Bales, K.R., Dodel, R.C., Liu, X., Glinn, M.A., Horn, J.W., et al., 1998. Alpha2-macroglobulin attenuates beta-amyloid peptide 1–40 fibril formation and associated neurotoxicity of cultured fetal rat cortical neurons. J. Neurochem. 70 (3), 1182–1188.

Dumoulin, M., Last, A.M., Desmyter, A., Decanniere, K., Canet, D., Larsson, G., et al., 2003.A camelid antibody fragment inhibits the formation of amyloidfibrils by human lyso-zyme. Nature 424 (6950), 783–788.

Fleming, A., 1922.On a remarkable bacteriolytic element found in tissues and secretions. Proc. R. Soc. Lond. B Biol. Sci. 93, 306–317.

Ganz, T., 2004.Antimicrobial polypeptides. J. Leukoc. Biol. 75 (1), 34–38.

Gupta, D.K., Theisen, N., von Figura, K., Hasilik, A., 1985.Comparison of biosynthesis and subcellular distribution of lysozyme and lysosomal enzymes in U937 monocytes. Biochim. Biophys. Acta 847 (2), 217–222.

Halim, A., Brinkmalm, G., Ruetschi, U., Westman-Brinkmalm, A., Portelius, E., Zetterberg, H., et al., 2011.Site-specific characterization of threonine, serine, and tyrosine glyco-sylations of amyloid precursor protein/amyloid beta-peptides in human cerebrospi-nalfluid. Proc. Natl. Acad. Sci. U. S. A. 108 (29), 11848–11853.

Halle, A., Hornung, V., Petzold, G.C., Stewart, C.R., Monks, B.G., Reinheckel, T., et al., 2008.

The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat. Immunol. 9 (8), 857–865.

Hansson, O., Zetterberg, H., Buchhave, P., Londos, E., Blennow, K., Minthon, L., 2006. Asso-ciation between CSF biomarkers and incipient Alzheimer's disease in patients with mild cognitive impairment: a follow-up study. Lancet Neurol. 5 (3), 228–234.

Hardy, J., Selkoe, D.J., 2002.The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297 (5580), 353–356.

Heneka, M.T., O'Banion, M.K., 2007.Inflammatory processes in Alzheimer's disease. J. Neuroimmunol. 184 (1–2), 69–91.

Hesse, C., Rosengren, L., Vanmechelen, E., Vanderstichele, H., Jensen, C., Davidsson, P., et al., 2000.Cerebrospinalfluid markers for Alzheimer's disease evaluated after acute is-chemic stroke. J. Alzheimers Dis. 2 (3–4), 199–206.

Hirth, F., 2010.Drosophila melanogaster in the study of human neurodegeneration. CNS Neurol. Disord. Drug Targets 9 (4), 504–523.

Hällgren, R., Terent, A., Venge, P., 1982.Lactoferrin, lysozyme, and beta 2-microglobulin levels in cerebrospinalfluid: differential indices of CNS inflammation. Inflammation 6 (3), 291–304.

Höltta, M., Hansson, O., Andreasson, U., Hertze, J., Minthon, L., Nagga, K., et al., 2013. Eval-uating amyloid-beta oligomers in cerebrospinalfluid as a biomarker for Alzheimer's disease. PLoS One 8 (6), e66381.

Jahn, T.R., Kohlhoff, K.J., Scott, M., Tartaglia, G.G., Lomas, D.A., Dobson, C.M., et al., 2011.

Detection of early locomotor abnormalities in a Drosophila model of Alzheimer's dis-ease. J. Neurosci. Methods 197 (1), 186–189.

Jonson, M., Pokrzywa, M., Starkenberg, A., Hammarstrom, P., Thor, S., 2015.Systematic Abeta analysis in Drosophila reveals high toxicity for the 1–42, 3–42 and 11–42 pep-tides, and emphasizes N- and C-terminal residues. PLoS One 10 (7), e0133272.

Kaplan, E.L., Meier, P., 1958.Nonparametric estimation from incomplete observations. J. Am. Stat. Assoc. 53 (282), 457–481.

Khabirova E. Cambridge: University of Cambridge; 2012.

Kitazawa, M., Oddo, S., Yamasaki, T.R., Green, K.N., LaFerla, F.M., 2005. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer's disease. J. Neurosci. 25 (39), 8843–8853.

Koch, H.P., Kurokawa, T., Okochi, Y., Sasaki, M., Okamura, Y., Larsson, H.P., 2008.

Multimeric nature of voltage-gated proton channels. Proc. Natl. Acad. Sci. U. S. A. 105 (26), 9111–9116.

Kumita, J.R., Helmfors, L., Williams, J., Luheshi, L.M., Menzer, L., Dumoulin, M., et al., 2012.

Disease-related amyloidogenic variants of human lysozyme trigger the unfolded pro-tein response and disturb eye development in Drosophila melanogaster. FASEB J. 26 (1), 192–202.

Lee, M., Kovacs-Nolan, J., Yang, C., Archbold, T., Fan, M.Z., Mine, Y., 2009.Hen egg lyso-zyme attenuates inflammation and modulates local gene expression in a porcine model of dextran sodium sulfate (DSS)-induced colitis. J. Agric. Food Chem. 57 (6), 2233–2240.

Lesne, S.E., Sherman, M.A., Grant, M., Kuskowski, M., Schneider, J.A., Bennett, D.A., et al., 2013.Brain amyloid-beta oligomers in ageing and Alzheimer's disease. Brain 136 (Pt 5), 1383–1398.

Lin, D.M., Goodman, C.S., 1994.Ectopic and increased expression of Fasciclin II alters mo-toneuron growth cone guidance. Neuron 13 (3), 507–523.

Liu, H., Zheng, F., Cao, Q., Ren, B., Zhu, L., Striker, G., et al., 2006.Amelioration of oxidant stress by the defensin lysozyme. Am. J. Physiol. Endocrinol. Metab. 290 (5), E824–E832.

Luo, J., Warmlander, S.K., Graslund, A., Abrahams, J.P., 2013.Human lysozyme inhibits the in vitro aggregation of Abeta peptides, which in vivo are associated with Alzheimer's disease. Chem. Commun. 49 (58), 6507–6509.

Luo, J., Warmlander, S.K., Graslund, A., Abrahams, J.P., 2014.Non-chaperone proteins can inhibit aggregation and cytotoxicity of Alzheimer amyloid beta peptide. J. Biol. Chem. 289 (40), 27766–27775.

(13)

Matarin, M., Salih, D.A., Yasvoina, M., Cummings, D.M., Guelfi, S., Liu, W., et al., 2015.A genome-wide gene-expression analysis and database in transgenic mice during de-velopment of amyloid or tau pathology. Cell Rep. 10 (4), 633–644.

Matsubara, E., Soto, C., Governale, S., Frangione, B., Ghiso, J., 1996.Apolipoprotein J and Alzheimer's amyloid beta solubility. Biochem. J. 316 (Pt 2), 671–679.

McGeer, P.L., McGeer, E.G., 2007.NSAIDs and Alzheimer disease: epidemiological, animal model and clinical studies. Neurobiol. Aging 28 (5), 639–647.

Montine, T.J., Phelps, C.H., Beach, T.G., Bigio, E.H., Cairns, N.J., Dickson, D.W., et al., 2012.

National Institute on Aging-Alzheimer's Association guidelines for the neuropatho-logic assessment of Alzheimer's disease: a practical approach. Acta Neuropathol. 123 (1), 1–11.

Nixon, R.A., Wegiel, J., Kumar, A., Yu, W.H., Peterhoff, C., Cataldo, A., et al., 2005.Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study. J. Neuropathol. Exp. Neurol. 64 (2), 113–122.

Nixon, R.A., Yang, D.S., 2011.Autophagy failure in Alzheimer's disease–locating the prima-ry defect. Neurobiol. Dis. 43 (1), 38–45.

Ogundele, M.O., 1998.A novel anti-inflammatory activity of lysozyme: modulation of serum complement activation. Mediat. Inflamm. 7 (5), 363–365.

Pahlsson, P., Shakin-Eshleman, S.H., Spitalnik, S.L., 1992.N-linked glycosylation of beta-amyloid precursor protein. Biochem. Biophys. Res. Commun. 189 (3), 1667–1673.

Portelius, E., Bogdanovic, N., Gustavsson, M.K., Volkmann, I., Brinkmalm, G., Zetterberg, H., et al., 2010.Mass spectrometric characterization of brain amyloid beta isoform signa-tures in familial and sporadic Alzheimer's disease. Acta Neuropathol. 120 (2), 185–193.

Qiao, X., Cummins, D.J., Paul, S.M., 2001.Neuroinflammation-induced acceleration of amyloid deposition in the APPV717F transgenic mouse. Eur. J. Neurosci. 14 (3), 474–482.

Raman, B., Ban, T., Sakai, M., Pasta, S.Y., Ramakrishna, T., Naiki, H., et al., 2005. AlphaB-crystallin, a small heat-shock protein, prevents the amyloidfibril growth of an amy-loid beta-peptide and beta2-microglobulin. Biochem. J 392 (Pt 3), 573–581.

Schwagerl, A.L., Mohan, P.S., Cataldo, A.M., Vonsattel, J.P., Kowall, N.W., Nixon, R.A., 1995.

Elevated levels of the endosomal-lysosomal proteinase cathepsin D in cerebrospinal fluid in Alzheimer disease. J. Neurochem. 64 (1), 443–446.

Sheng, J.G., Bora, S.H., Xu, G., Borchelt, D.R., Price, D.L., Koliatsos, V.E., 2003. Lipopolysac-charide-induced-neuroinflammation increases intracellular accumulation of amyloid precursor protein and amyloid beta peptide in APPswe transgenic mice. Neurobiol. Dis. 14 (1), 133–145.

Wogulis, M., Wright, S., Cunningham, D., Chilcote, T., Powell, K., Rydel, R.E., 2005.

Nucleation-dependent polymerization is an essential component of amyloid-mediated neuronal cell death. J. Neurosci. 25 (5), 1071–1080.

Wyss-Coray, T., Rogers, J., 2012.Inflammation in Alzheimer disease-a brief review of the basic science and clinical literature. Cold Spring Harb. Perspect. Med. 2 (1), a006346.

Yazaki, M., Tagawa, K., Maruyama, K., Sorimachi, H., Tsuchiya, T., Ishiura, S., et al., 1996.

Mutation of potential N-linked glycosylation sites in the Alzheimer's disease amyloid precursor protein (APP). Neurosci. Lett. 221 (1), 57–60.

Yerbury, J.J., Kumita, J.R., Meehan, S., Dobson, C.M., Wilson, M.R., 2009. alpha2-Macroglobulin and haptoglobin suppress amyloid formation by interacting with prefibrillar protein species. J. Biol. Chem. 284 (7), 4246–4254.

References

Related documents

CSF biomarkers in Alzheimer’s disease (AD) include β-amyloid 42 (Aβ42), total-tau (T-tau) and phosphorylated-tau (P-tau), reflecting brain amyloid, axonal and tangle

Abbreviations Aβ, β amyloid AD, Alzheimer’s disease ADAM, A Disintegrin And Metalloproteinase APOE, Apolipoprotein E APP, amyloid precursor protein AUROC, area under the

Delineating amyloid plaque associated neuronal sphingolipids in transgenic Alzheimer's disease mice (tgArcSwe) using MALDI imaging mass spectrometry.. Zanni G, Michno W, Di Martino

Michno W, Nyström S, Wehrli P, Lashley T, Brinkmalm G, Guerard L, Syvänen S, Sehlin D, Kaya I, Brinet D, Nilsson KPR, Hammarström P, Blennow K, Zetterberg H, Hanrieder

The results (Figure 3, A and B) demonstrate that 1 nM fibrillar Aβ40 significantly induced H3 acetylation and phosphorylation and furthermore, that 10 nM oligomeric

In paper III, we investigated genetic variation in the purinergic P2Y12 gene in a case-control study and found a haplotype to be associated with increased risk of AD. In

Brain beta-amyloid measures and magnetic resonance imaging atrophy both predict time-to-progression from mild cognitive impairment to Alzheimer’s disease.. Defining imaging

[r]