prion 2019 emerging concepts - edinburgh research explorer

143
Edinburgh Research Explorer Optimization of RT-QuIC for detection of seeding activity in preclinical blood samples from prion-infected sheep. Citation for published version: Thomas, C, McCutcheon, S, Alejo-Blanco, R, Tan, BC & Houston, F 2019, 'Optimization of RT-QuIC for detection of seeding activity in preclinical blood samples from prion-infected sheep.', Prion 2019, Edmonton, Canada, 21/05/19 - 24/05/19 pp. 1–141. https://doi.org/10.1080/19336896.2019.1615197 Digital Object Identifier (DOI): 10.1080/19336896.2019.1615197 Link: Link to publication record in Edinburgh Research Explorer Document Version: Publisher's PDF, also known as Version of record General rights Copyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorer content complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 12. Sep. 2022

Upload: khangminh22

Post on 07-May-2023

1 views

Category:

Documents


0 download

TRANSCRIPT

Edinburgh Research Explorer

Optimization of RT-QuIC for detection of seeding activity inpreclinical blood samples from prion-infected sheep.

Citation for published version:Thomas, C, McCutcheon, S, Alejo-Blanco, R, Tan, BC & Houston, F 2019, 'Optimization of RT-QuIC fordetection of seeding activity in preclinical blood samples from prion-infected sheep.', Prion 2019, Edmonton,Canada, 21/05/19 - 24/05/19 pp. 1–141. https://doi.org/10.1080/19336896.2019.1615197

Digital Object Identifier (DOI):10.1080/19336896.2019.1615197

Link:Link to publication record in Edinburgh Research Explorer

Document Version:Publisher's PDF, also known as Version of record

General rightsCopyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s)and / or other copyright owners and it is a condition of accessing these publications that users recognise andabide by the legal requirements associated with these rights.

Take down policyThe University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorercontent complies with UK legislation. If you believe that the public display of this file breaches copyright pleasecontact [email protected] providing details, and we will remove access to the work immediately andinvestigate your claim.

Download date: 12. Sep. 2022

Full Terms & Conditions of access and use can be found athttps://www.tandfonline.com/action/journalInformation?journalCode=kprn20

Prion

ISSN: 1933-6896 (Print) 1933-690X (Online) Journal homepage: https://www.tandfonline.com/loi/kprn20

PRION 2019 emerging concepts

To cite this article: (2019) PRION 2019 emerging concepts, Prion, 13:sup1, 1-141, DOI:10.1080/19336896.2019.1615197

To link to this article: https://doi.org/10.1080/19336896.2019.1615197

© 2019 The Author(s). Published by InformaUK Limited, trading as Taylor & FrancisGroup.

Published online: 18 May 2019.

Submit your article to this journal

Article views: 12614

View related articles

View Crossmark data

Citing articles: 2 View citing articles

PRION 2019 emerging concepts

Co- Editors:David WestawayCentre for Prions and Protein misfolding Diseases,University of Alberta

Hermann SchaetzlCalgary Prion Research Unit, University of Calgary

Kevin KeoughAlberta Prion Research Institute, Alberta Innovates

Dear attendees,

Welcome to Prion 2019 in downtown Edmonton.

With over 320 participants from 25 countries, we hopeyou are greatly stimulated by this conference. Over thecoming days, 47 oral presentations, over 200 posters,two workshops and three special sessions will delivercutting-edge concepts to understand and control debil-itating prion and related neurodegenerative diseases.We hope you make the acquaintance of new theories,new technologies and - most importantly - new collea-gues and collaborators.

We thank all our co-organizers and we wish you awarm welcome from the province nd people of Alberta.

David, Hermann and Kevin

1. Interspecies transmission of the chronic wastingdisease agent

Justin Greenlee

Virus and Prion Research Unit, National Animal Disease Center,USDA Agriculture Research Service

ABSTRACT

The presentation will summarize the results of variousstudies conducted at our research center that assess thetransmissibility of the chronic wasting disease (CWD)agent to cattle, pigs, raccoons, goats, and sheep. Thiswill include specifics of the relative attack rates, clinicalsigns, and microscopic lesions with emphasis on how todifferentiate cross-species transmission of the CWDagent from the prion diseases that naturally occur inhosts such as cattle or sheep. Briefly, the relative

difficulty of transmitting the CWD agent to sheep andgoats will be contrasted with the relative ease of trans-mitting the scrapie agent to white-tailed deer.

2. RT-QuIC seed amplification assays in thediagnosis of prion diseases, synucleinopathies andtauopathies

Byron Caughey

Senior Investigator at Rocky Mountain Laboratories, NIAID, NIH

ABSTRACT

Antemortem diagnoses of many proteopathies can bedifficult, due in part to an inability to detect specificcausative misfolded protein aggregates with sufficientsensitivity, specificity and practicality. To address thisissue we and others have developed RT-QuIC assaysthat exploit the ability of such aggregates to seed thepolymerization of protein monomers into amyloidfibrils. PrP-based RT-QuIC assays for are available forvirtually all prion diseases of mammals and have beenadapted to multiple specimens including, most recently,the skin and eyes of CJD cases. Analyses of CSF andnasal brushings can give nearly 100% accurate ante-mortem diagnosis of sCJD. We and others have adaptedthe seed amplification approach to the detection ofsynucleinopathies such as Parkinson’s disease. αSynRT-QuIC analyses of patients’ CSF can detect α-synu-clein seeds early in the clinical phase of disease and hasprovided unprecedented diagnostic accuracy. We havealso developed three types of ultrasensitive tau RT-QuIC assays optimized for (i) Alzheimer disease andchronic traumatic encephalopathy (i.e., 3R/4R tauopa-thies), (ii) Pick disease (a 3R tauopathy), or (iii) pro-gressive supranuclear palsy and other 4R tauopathies.Each assay provides strong selectivity for the type oftauopathy for which it was optimized. Such selectivitiesmay underpin the propagation of specific tau aggregateconformers in vivo. In summary, these assays can facil-itate diagnosis and allow measurements of proteopathicbiomarkers. This, in turn, may promote the therapeutictrials by improving patient cohort selection and long-itudinal assessments of drug efficacy.

This article has been republished with minor changes. These changes do not impact the academic content of the article.

PRION2019, VOL. 13, NO. S1, 1–141https://doi.org/10.1080/19336896.2019.1615197

© 2019 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricteduse, distribution, and reproduction in any medium, provided the original work is properly cited.

3. A perspective on prion strains and theirsignificance

John Collinge

Department of Neurodegenerative Disease, Institute of Neurology,University College London (UCL)

ABSTRACT

Mammalian prions are thought to comprise multimericassemblies of misfolded cellular prion protein (PrPC)which propagate by recruitment of PrP monomers andsubsequent fission. Although devoid of a nucleic acidgenome, mammalian prions exist as multiple strainswhich can be serially propagated in laboratory animalsand produce distinct patterns of disease. Since multiplestrains can be maintained in the same inbred mouse linewith identical PrP genes, strains cannot be encoded bydifferences in the primary structure of PrP and it isthought they represent structurally distinct seeds thatare able to recruit host PrPC into their distinct polymericforms. These strain-specific biological and structuralproperties can be maintained even following passagethrough an intermediate mammalian species with a dif-ferent PrP primary structure. In addition to the funda-mental biological interest in such protein-basedinheritance, it is clear that understanding the prion strainphenomenon is fundamental to understanding prion dis-ease pathogenesis and transmission properties. Critically,prion strains, rather than being a biological clone, appearto constitute a cloud of diverse molecular assemblies orquasispecies which can adapt and mutate under host andother selection pressures. This has an important bearingon therapeutic strategies which need to avoid the devel-opment of drug resistance. A proof of principle of target-ing neuronal PrPC, rather than disease-associatedassemblies, was established and our therapeutic approachusing an anti-human PrPC monoclonal antibody inpatients with CJD is underway. Prion strains may beconsidered as subtypes of misfolded PrP assemblies withthe necessary properties and replication kinetics to evadecellular and other host defences, propagate exponentiallyand be able to act as an efficient pathogen, while otherpolymeric PrP forms are degraded or form highly stableaggregates that do not propagate and/or are relativelyinert. The very diversity of prion quasispecies, the degreeof which may itself vary between strains, may be funda-mental to adaptation to a new host (and to particulartissues within it) to allow its survival and spread. Thismay also account for why some amyloids are infectiouspathogens and others not and understanding the struc-tural basis of these different pathobiological propertieswill be of major importance.

4. A unified model of prion infectivity and strainmaintenance by cofactor selection

Surachai Supattapone

Departments of Biochemistry and Medicine, Dartmouth College

ABSTRACT

The protein-only hypothesis predicts that infectiousmammalian prions are composed solely of PrPSc, a mis-folded conformer of the normal prion protein, PrPC.However, to date, all wild-type protein-only PrPSc pre-parations lack significant levels of prion infectivity. Usinga systemic biochemical approach, our laboratory isolatedand identified two different endogenous cofactor mole-cules, RNA and phosphatidylethanolamine, which facil-itate the formation of prions with high levels of specificinfectivity (>106-fold greater than protein-only PrPSc),leading us to propose to the alternative hypothesis thatcofactor molecules are required to form wild-type infec-tious prions. In addition, we found that purified cofactormolecules restrict the strain properties of chemically-defined infectious prions[4], suggesting a “cofactor selec-tion”model in which natural variation in the distributionof strain-specific cofactor molecules in different parts ofthe brain may be responsible for strain-dependent pat-terns of neurotropism. Recently, we have discovered thatprions with parental strain properties and full specificinfectivity can be restored from non-infectious protein-only PrPSc in vitro (Burke, et al. PLoS Pathogens, in press).The restoration reaction is rapid, potent, and requiresbank vole PrPC substrate, post-translational modifica-tions, and cofactor molecules. These findings provideevidence for a unified model of prion infectivity inwhich the global structure of protein-only PrPSc canaccurately store latent infectious and strain information,but cofactor molecules control a reversible switch that canunmask full biological infectivity. The results are alsoconsistent with the idea that prion strains select compa-tible cofactor molecules on the basis of their ability tomaintain strain-specific PrPSc conformations.

5. Structural insights into the mechanism ofmammalian prion propagation

Witold K. Surewicz

Department of Physiology and Biophysics, Case Western ReserveUniversity

ABSTRACT

Despite recent progress in structural studies of orderedprotein aggregates such as amyloid fibrils in general,

2 ABSTRACT

the structure of infectious mammalian prions is poorlyunderstood and controversial. This presentation willdiscuss structural aspects of prion propagation, with aspecial focus on a model system of PrP23-144 amyloidthat can reproduce in vitro the phenomena of strainmultiplicity and transmissibility (seeding) barriers andis amenable to high-resolution structural characteriza-tion. Furthermore, we will discuss mass spectrometry-based approaches that can be used for structural char-acterization of brain-derived prions and the insightsprovided by these studies regarding the structuralbasis of prion infectivity.

6. Seed-induced Aβ deposition impairs adultneurogenesis in mouse models of Alzheimer’sdisease

Meyer-Luehmann

Department of Neurology, Medical Center-University of Freiburg,Germany and Faculty of Medicine, University if Freiburg, Germany

ABSTRACT

Alzheimer´s disease (AD) is characterized by severe neu-ronal loss as well as the accumulation of amyloid-β (Aβ)which ultimately leads to plaque formation. These Aβdeposits can be induced in young pre-depositing APPtransgenicmice by intracerebral injection of Aβ-containingbrain homogenate. Although a decline of neurogenic capa-city in the brain of AD patients and ADmouse models hasbeen reported, our understanding of whether this impair-ment is specifically altered by Aβ plaques is limited. Here,we find that induced Aβ deposition (Aβ seeding), repre-senting early stages of plaque formation, leads to a dramaticdecrease in adult neurogenesis in the hippocampus andolfactory bulb. We further report that the generation andmaturation of newborn neurons is affected by the induc-tion of Aβ deposition in both neurogenic niches. Notably,cell death and apoptosis occur in conjunction with Aβseeding and the exposure to enriched environment andvoluntary running reduces Aβ seeding via activated micro-glia and vivifies neurogenesis and proliferation.

7. Modifying prion spread through the CNS

Christina Sigurdson

Department of Pathology, University of California, San Diego

ABSTRACT

Prion aggregates increase exponentially and rapidlytransit through the central nervous system duringprion disease, yet the driving factors from the hostand the prion that ultimately govern spread are unclear.

Using a series of mouse models with alterations in theextracellular matrix or in the prion conformation, weinvestigated the determinants of prion propagationthrough the brain. By targeting heparan sulfate bio-synthesis, we find that prion deposition sites shift andthe survival time is prolonged in a strain dependentmanner. These findings suggest that manipulatingendogenous co-factors may be exploited to enhanceprion clearance.

8. Drivers of neurotoxicity in prion diseases

Adriano Aguzzi

Director, Institute of Neuropathology, University of Zurich

ABSTRACT

While the mechanics of prion replication have beensubjected to intense studies and are becoming increas-ingly clear, little is known about what ultimatelycauses brain damage. It cannot just be mechanicaldamage from the accumulation of PrPSc, because thelatter is present only in trace amounts – in contrast tosystemic amyloidoses with enormous amyloid loads ine.g. spleen, liver or peripheral nerves. This suggests theexistence of specific pathways of brain damage. If suchpathways exist, their components may be exploitableas targets of therapeutic intervention. My lab hasfocused on the latter question using many differentapproaches. For example, we have studied themechanism of action of neurotoxic antibodies againstPrP. One such antibody (denominated POM1) wasfound to be a prion-mimetic, which elicits the samecellular programmes as bona fide prion infections, andcan be antagonized by compounds which protectagainst prion toxicity. Currently, we are exploringthe biogenesis of spongiform changes, arguably themost typical consequence of prion disease. It turnsout that neuronal vacuolation, the basis of spongiformchanges, is controlled by cellular checkpoint moleculesthat are subverted by prion infection and by prion-mimetic antibodies. The nature of said checkpointswill be discussed.

9. Targeting proteopathic seeds in Alzheimer´sdisease

Mathias Jucker

Department of Cellular Neurology, Hertie Institute for Clinical BrainResearch, University of Tübingen, and German Center forNeurodegenerative Diseases (DZNE), D-72076 Tübingen, Germany

PRION 3

10. Prion accumulation in the bone marrow: theorigin of prionemia?

Olivier Andreolettia, Jean-Yves Douetb, Alvina Huorb,Séverine Luganb, Naima Aronb, Cécile Tillierb, HervéCassardb and Olivier Andreolettib

aINRA Research Director, UMR INRA ENVT 1225; bUMR INRA ENVT1225, Interactions Hôtes Agents Pathogènes, 23 chemin descapelles 31076 Toulouse, FRANCE

ABSTRACT

The presence of significant amount of Prion infectivityin the bone marrow (BM) of both sCJD and vCJDaffected patients has been demonstrated. This raisedquestions about the role of BM in the occurrence ofprionemia and the risk of disease transmission asso-ciated with this tissue. To address these questions, wedeveloped a bone marrow transplantation model inPrP+/+ and PrP0/0 C57Bl6 mice. Despite the presenceof significant amount of infectivity in the bone marrowof RML infected PrP +/+ C57Bl6 mice, the reconstitu-tion of sub-lethally γ-irradiated mice (8.5 Gy) with thistissue failed to transmit Prion disease or to cause adetectable prionemia. The RML peripheral inoculationof irradiated and BM reconstituted mice clearly demon-strated that the prion replication capacity of the radio-sensitive BM cell populations has no detectable impacton the occurrence and the level of Prionemia. It alsoindicated that the radio resistant cells compartment isassociated with the vast majority of the infectivity in theBM. In RML inoculated LT−/- / TNFα −/- C57Bl6, prionaccumulated in Bone Marrow tissue but mice displayeda very low/undetectable prionemia. Together theseresults support the contention that conversely to BM,germinal lymphoid centers play a central role in theoccurrence and the level of prionemia.

11. Arguments for Alzheimer’s and Parkinson’sdiseases caused by prions

Stanley B. Prusiner

Institute of Neurodegenerative Diseases, and Professor of Neurologyand Biochemistry, University of California San Francisco

ABSTRACT

Arguments for Alzheimer’s (AD) and Parkinson’s dis-eases (PD) being caused by prions continue to advancewith new evidence. Findings in the brains of deceasedAD patients argue that both Aβ and tau prions can bedemonstrated by bioassays in cultured cells as well as intransgenic (Tg) mice. Likewise, studies of the brains of

deceased MSA patients have been found to contain α-synuclein prions by bioassays in cultured cells and Tgmice. Conversely, the brains of AD patients do notcontain α-synuclein prions, and the brains of MSApatients do not contain Aβ or tau prions.Additionally, while the brains of patients who died ofeither progressive supranuclear palsy (PSP) or cortico-basal degeneration (CBD) contained tau prions, neitherAβ nor α-synuclein prions were detectable. Merelymeasuring the levels of Aβ, tau, and α-synucleinappears to give misleading information about the etiol-ogy and pathogenesis of neurodegenerative diseases(NDs). From a large array of bioassays, we concludethat AD, PD, MSA, and the frontotemporal dementias,including PSP and CBD, are all prion diseases. Ourfindings argue that changes in the conformations ofAβ, tau, and α-synuclein underlie the acquisition ofprion infectivity in all of these NDs.

12. Genetic risk factors for sporadic CJD:replication, expression, function

Simon Mead

UCL Institute of Prion Diseases and NIHR Senior Investigator

ABSTRACT

By definition, sporadic Creutzfeldt-Jakob disease is notcaused by genetic mutation of the prion protein gene,and the patient has no identifiable environmental expo-sure. Like common neurodegenerative dementias, age isa powerful risk factor, but the molecular mechanisms ofincreasing risk with advanced age are unclear. Weresearch both genomic and epigenomic factors inhuman prion disease by genome-wide associationstudy. Last year we completed initial analysis of genomewide association study with contributions of patientsamples from UK, USA, Germany, France, Italy,Netherlands, Austria, Spain, Switzerland andAustralia. We have now analysed over 5,000 case sam-ples and a large number of controls. Additional toPRNP, we identified other genetic loci at which multi-ple SNPs surpass statistical thresholds for genome-widesignificance. A SNP at one of these loci, near to STX6,is already known to be a genetic risk factor for atauopathy, progressive supranuclear palsy. We havefollowed up these findings by replication, investigationof correlations with clinical phenotypes, allelic expres-sion of genes in blood and brain and in experimentalmodels. Genetic risk factors point to critical aspects ofthe pathobiology of prion disease and should beassessed for their potential as therapeutic targets.

4 ABSTRACT

13. A solid-state of conceptualization ofinformation transfer from gene to message protein

Steve McKnight

Department of Biochemistry UT Southwestern

ABSTRACT

My presentation will describe speculative ideas andearly stage research concerning the flow of geneticinformation from the nuclear residence of genes tothe disparate, cytoplasmic sites of protein synthesis.This process is particularly important for neurons, inwhich the nuclear location of genes may be far-removed from localized sites of mRNA translation atactive synapses. I propose that this process of informa-tion transfer is meticulously guided by transient struc-tures formed from protein segments of low sequencecomplexity or intrinsic disorder. These low complexitydomains are ubiquitously associated with regulatoryproteins that control gene expression and RNA biogen-esis, but they are also found in the central channel ofnuclear pores, the nexus points of intermediate filamentassembly, and the locations of action of other well-studied cellular proteins and pathways. Upon beingorganized into localized cellular positions via mechan-isms utilizing properly folded protein domains, therebyfacilitating elevated local concentration, certain lowcomplexity domains adopt cross-β interactions asdescribed 60-70 years ago by Linus Pauling.Derivation of the atomic structure of labile polymersformed from the low complexity domain of the fused insarcoma (FUS) RNA binding protein has demonstratedhow these cross-β interactions can be both structurallyspecific and labile to disassembly. These weakly teth-ered assemblies, I propose, are built to relay the passageof genetic information from one site to another withina cell, ensuring that the process is of extreme fidelity.

14. Beyond neurofibrillary tangles in tauopathy

Karen Hsiao Ashe and N. Bud Grossman

Center for Memory Research and Care, Edmund Wallace and AnneMarie Tulloch Chairs in Neurology and Neuroscience at theUniversity of Minnesota

ABSTRACT

Soluble forms of tau, unrelated to neurofibrillary tan-gles, play pathogenic roles in tauopathies. A soluble taufragment, Δtau314 that resists forming fibrils formswhen caspase-2 (CASP2) cleaves human tau atAsp314. Δtau314 is elevated in forebrain tissue of

humans with mild cognitive impairment, Alzheimer'sdisease, Huntington's disease and Lewy bodydementia. This cleavage event, along with tau phos-phorylation, leads to tau mislocalization and reducedAMPA receptor levels in dendritic spines and abnormalpost-synaptic function (reduced mEPSCs). Renderingendogenous mouse tau (mTau) non-cleavable byCASP2 by introducing a homologous mutation inmTau, Asp303-to-Glu reduces mTau in dendriticspines, increases excitatory synaptic transmissionand resistance to excitotoxin-induced seizures, andameliorates premature mortality in transgenic miceexpressing human amyloid precursor protein. Thesedata support a role for non-amyloid forms of proteinspresent in the neuropathological amyloid-containingstructures (e.g., tangles) that characterize their respec-tive diseases, and suggest a strategy for repairing synap-tic transmission in tauopathies by inhibiting CASP2.

15. Cellular prion infection: from traffic jams tonew drug targets

Sabine Gilch

Canada Research Chair in Prion Disease Research, University ofCalgary

ABSTRACT

Prion infection on a cellular level is characterized bythe presence of PrPSc aggregates at the plasma mem-brane, in vesicles along the endo-lysosomal pathwayand in recycling endosomes, as well as an increasedlevel of unesterified cholesterol. High cholesterol con-tent can affect membrane properties and vesicle traf-ficking. Our goal is to understand the impact of prioninfection on endo-lysosomal vesicle trafficking, todefine a link with aberrant cholesterol metabolismand to validate new therapeutic compounds that targetthose pathways. We show that prion infection induces a‘traffic jam’ by impairing with retrograde cargo trans-port and lysosomal degradation capacity, processes thatrequire functions of the retromer complex and rab7,respectively. We show that stimulating retrogradetransport, lysosomal degradation capacity and enhan-cing cholesterol efflux in neuronal cells reduces prionpropagation. Overall, our studies provide new insightsinto prion-induced cellular dysfunction. We observedalterations similar to those described in Alzheimer’sdisease, indicating that the endo-lysosomal systemmight be an intersection in the pathogenesis of thoseneurodegenerative diseases.

PRION 5

16. Design and optimization of nucleic acidderivatives for inhibition of prion-like propagationat W32 site of SOD1 enzyme in Amyotrophic LateralSclerosis

Vijaya Kumar Hingea,b, Nikolay Blinova,b, DipankarRoya,b, W. Ted Allisona, David S. Wisharta and AndriyKovalenkoa,b

aUniversity of Alberta, Edmonton, Canada; bNanotechnologyResearch Centre, Edmonton, Canada

CONTACT Vijaya Kumar Hinge [email protected]

ABSTRACT

Misfolding of human Cu/Zn superoxide dismutase(SOD1) cytosolic metalloenzyme results in the forma-tion of neurotoxic oligomers in familial AmyotrophicLateral Sclerosis (fALS). Over 180 mutations have beenidentified and linked to the gene encoding of SOD1 infALS. Currently, there is no cure for ALS and only twoFood and Drug Administration (FDA)-approved drugs,Riluzole and Edaravone, are available for treatment ofALS patients. The human cell culture studies demon-strated that misfolded SOD1 propagates from cell tocell in a prion-like fashion and W32 residue is impor-tant for templated misfolding of wild type SOD1(wtSOD1) into toxic aggregates [1]. The recent animalstudies on ALS Zebrafish embryo model suggest thatmutation of W32S decreases the toxicity to motor neu-ron functions [2]. The inhibition of pathological mis-folding of SOD1 via drug-like molecules binding atW32 site is a novel and promising therapeutic strategyfor the development of ALS therapy.

In silico de novo drug design and pharmacophore-restrained high-throughput virtual screening (HTVS)methods were used to identify the anti-ALS lead candidatesat W32 site. A library of commercially available lead-likecompounds (650,000) and Food and Drug Administration(FDA) approved small molecules were screened at W32site of SOD1 using HTVS and were filtered through auracil-based pharmacophore model. Based on the analysisof physico-chemical descriptors and pharmacophore fea-tures of the experimental binding mode of 5-FUrd atW32,a group of new Anti-ALS lead compounds were rationallydesigned. Anti-ALS lead candidates were ranked with in-house quantitative structure–activity relationship (QSAR)models based on the 3D-RISM-KH molecular solvationtheory derived molecular descriptors which accurately pre-dicted the blood brain barrier (BBB) permeability [3]. Anew protocol was developed to account for structural sol-vation effects and was successfully validated via predictingexperimental binding modes of 5-FUrd at W32 site of

SOD1. The protocol combines the water placement algo-rithm based on the 3D-RISM-KH molecular theory ofsolvation and molecular docking simulations implementedin the Molecular Operating Environment (MOE) inte-grated drug discovery package. One of the anti-ALS leadcompound, Telbivudine, tested in ALS-Zebrafish embryomodel significantly rescued axonopathy in a dose-depen-dent manner [2,4]. The newly designed lead-like com-pounds were selected by virtual screening, and de novodrug design methods show higher affinity compared tothe Telbivudine and can potentially inhibit prion-like pro-pagation of misfolding of SOD1.

References

[1] Grad L, Guest WC, Yanai A, et al. Intermolecular trans-mission of superoxide dismutase 1 misfolding in livingcells. Proc Natl Acad Sci USA. 2011;108:16,398–403.

[2] Duval et al. Neurobiology of disease 2019;124:297–310.[3] Roy et al. ACS Omega. 2019 (accepted).[4] Treatment of amyotrophic lateral sclerosis (ALS). Patent

application no 62/637,013. USPTO (pending); 2018.

17. Large-scale production of phospholipidcofactor recombinant prions with high specificinfectivity

Daniel J. Walsha, Marcus D. Tuttleb, Cassandra M.Burkea, Kurt W. Zilmb, and Surachai Supattaponea,d

aDepartments of Biochemistry and Cell Biology, Geisel School ofMedicine at Dartmouth, Hanover, New Hampshire, USA;bDepartment of Chemistry, Yale University, New Haven,Connecticut, USA; cMedicine, Geisel School of Medicine atDartmouth, Hanover, New Hampshire, USA

CONTACT Daniel J. Walsh [email protected]

ABSTRACT

The fundamental event underlying mammalian prioninfectivity is the induced conformational change of PrPC

into PrPSc. A major goal in the field is to determine thestructural mechanism of this conversion process, and acritical step towards this goal would be to determine theprotein structure of fully infectious PrPSc. One techniquethat could be used to determine the structure of infectiousPrPSc with high resolution is solid-state (ss)NMR, butapplication of this method requires the production of milli-gram quantities of a homogeneous, isotopically-labelledPrPSc sample whose specific infectivity is equivalent tothat of authentic brain-derived PrP27-30 (~106 LD50/µgPrP) [1].

We previously used sPMCA to produce two chemicallydefined recombinant (rec)PrPSc conformers originally

6 ABSTRACT

derived from the same seed. Whereas cofactor recPrPSc

produced with phospholipids displayed high levels of spe-cific infectivity, protein-only recPrPSc produced from PrPalone was completely non-infectious [2]. A limitation ofPMCA technology is that it can only be used to producemicrogram quantities of PrPSc, and therefore is not afeasible option for generating ssNMR samples. To over-come this limitation, we produced large quantities of cofac-tor recPrPSc and protein-only recPrPSc by shaking ratherthan PMCA. Shaking-produced cofactor recPrPSc causedscrapie in both wild-type mice and bank voles (homozy-gous M109), while protein-only recPrPSc failed to infecteither species. End-point titration bioassays indicated thatthe specific infectivity of shaking-produced cofactorrecPrPSc was ~106 LD50/µg PrP, while that of protein-only recPrPSc was <5 LD50/µg PrP. These results confirmthat shaking-produced phospholipid cofactor recPrPSc is agood candidate for structural studies of bona fide mamma-lian prion infectivity. The morphologies of cofactorrecPrPSc and protein-only recPrPSc molecules could notbe easily distinguished by negative-stain electron micro-scopy, but differences in the secondary structure betweenthe two conformers can be detected by ssNMR.

References

[1] Deleault NR, Harris BT, Rees JR, et al. Formation ofnative prions from minimal componenets in vitro. ProcNatl Acad Sci U S A. 2007;104(23):9741–9746.

[2] Deleault NR, Walsh DJ, Piro JR, et al. Cofactor mole-cules maintain infectious conformation and restrictstrain properties in purified prions. Proc Natl AcadSci U S A. 2012;109(28):E1938–E1946.

18. Targeting protein aggregation throughnanoparticle-based inhibitors

Bibin G. Ananda,b,c, Kailash Prasad Prajapathib, KritiDubeyb and karunakar Karb

aDepartment of Bioscience and Bioengineering, Indian Institute ofTechnology Jodhpur, India; bSchool of Life science JawaharlalNehru University, New Delhi, India; cCenter for Prions and Proteinfolding diseases, University of Alberta, Edmonton, Canada

Abstract not available.

19. Amplification of prions derived from singlecells isolated from different brain regions of RMLinfected animals by laser capture microdissection

D. Gorskia, A. Lyona, A. Mukherjeea, S. Medinab, S.Boothb, S. Pritzkowa and C. Sotoa

aMitchell Center for Alzheimer’s disease and related Brain disorders,University of Texas Medical School at Houston; bZoonotic Diseases

and Special Pathogens, National Microbiology Laboratory, PublicHealth Agency of Canada

CONTACT D. Gorski [email protected]

ABSTRACT

Prion diseases are uniformly fatal neurodegenerative dis-orders characterized by the misfolding and aggregation ofhost-encoded prion protein (PrPC) into a β-sheet rich,insoluble, and protease-resistant conformation termedPrPSc, which serves as the sole infectious agent.Depending on the prion disease, PrPSc can adopt differentisoforms, which preferentially deposit in distinct brainregions and differentially accumulate in the neurons,astroglia, and microglia of these areas. Currently, thecellular factors and processes governing the differentialcellular vulnerability to PrPSc deposition and damageamongst different cell types and brain regions are notknown. The putative differences in biochemical, biologi-cal, and structural characteristics of PrPSc associated withdifferent cells and regions is also yet to be described.Indeed, several previous reports have shown that differentprion strains can co-exist in the same brain and may bemasked by a single dominant pathological phenotype. Inthis study, we optimized the protein misfolding cyclicamplification (PMCA) technology to amplify PrPSc froma single cell. For these studies we used a cell line chroni-cally infected with RML prions and dilute it to add indi-vidual cells per sample. Serial dilutions of infected cellculture showed that PMCA enabled amplification ofprions from a single cell. This was further confirmed byplating cell dilutions. The presence of a single cell wasconfirmed via microscopy. We then applied this techni-que to amplify PrPSc from individual cells extracted fromthe brain of prion infected animals via laser capture dis-section. We show successful amplification of prions fromindividual neurons from some areas of the brain and arecurrently analysing the presence of PrPSc in neurons andglial cells extracted via LCM from the cerebellum, thala-mus, cortex, and hippocampus of RML infected mice.Partial fixation and staining of the tissue prior to LCMdid not interfere with PMCA amplification. This technol-ogy will enable to study the characteristics of prionsassociated with different cell types isolated from distinctareas of the brain of animals and humans infected withdifferent prion strains. Experiments can be done compar-ing PrPSc in neuronal versus glial cell types originatingfrom distinct brain regions as well distinct time points ofdisease progression. These findings can then be coupledwith single cell genomics and proteomics analysis. Thesefindings will shed light on the molecular basis of differ-ential cellular vulnerability to prion infection and thenerve tropism of distinct prion strains.

PRION 7

20. New insights into the structure of bovinespongiform encephalopathy (BSE) prions

Razieh Kamali-Jamila,b, Sara Amidiana,b, BrianTancownya,b, Xiongyao Wanga,b, Ester Vázquez-Fernándeza,b, Xinli Tanga, Howard Youngb andHolger Willea,b

aCentre for Prions and Protein Folding Diseases, University ofAlberta, Edmonton, Canada; bDepartment of Biochemistry, Facultyof Medicine & Dentistry, University of Alberta, Edmonton, Canada

CONTACT Razieh Kamali-Jamil [email protected]

Present address: Xiongyao Wang, School of MaterialsScience and Engineering, Harbin Institute of Technology,Weihai, Shandong, China

ABSTRACT

Background: Bovine spongiform encephalopathy (BSE)is caused by the conversion of the cellular prion protein(PrPC) to the infectious form (PrPSc). Prion diseases incattle include classical BSE (C-type BSE), and atypicalstrains known as H-type and C-type BSE. Consumptionof BSE-infected meat products may have caused variantCreutzfeldt-Jakob disease in humans. Previously, thestructure of the recombinant prion protein has beenanalysed using X-ray crystallography and NMR spec-troscopy methods. However, the PrPSc has hamperedall attempts at structural determination due to its inso-lubility and propensity to aggregate. A recent cryo-EMstudy on glycosylphosphatidylinositol-anchorless PrPSc

proposed a four-rung ß-solenoid architecture for indi-vidual PrPSc molecules. Currently, no three-dimen-sional structure is available for BSE prions. The aimof this project is the characterization and detailed ana-lysis of the structure of BSE prions.Materials and Methods: L-type BSE prions were pur-ified from the brains of prion-infected transgenic miceexpressing bovine PrPC, using phosphotungstate anionsfollowed by sucrose gradient centrifugation. The pur-ification quality was assessed using SDS PAGE followedby silver staining or Western blotting. The purifiedsamples were analysed via negative stain electronmicroscopy to determine the quality of the samplepreparations. We also developed an immunogold label-ling protocol for the specific detection of BSE fibrils onelectron microscopy grids. Next, electron micrographsof individual BSE fibrils were analysed through a vari-ety of image processing techniques in order to obtainthree-dimensional reconstructions. For this purpose,fibril images with a clear helical twist were selected,segmented, aligned, classified, and oriented.Subsequently, a three-dimensional volume could begenerated through back projection.

Results and Conclusion: The successful purification ofBSE prion fibrils was confirmed by Western blottingand negative stain EM. Image analyses revealed a het-erogeneous population of BSE fibrils consisting of twointertwined protofibrils, in agreement with the resultsof previous studies. Additionally, fibrils with only oneprotofilament were observed, which is a novel finding.2D class averages on more than 500 fibrils furtherconfirmed the two distinct morphologies of the L-typeBSE fibrils. We postulate that the two-protofilamentBSE fibrils result from the interaction of two individualfibrils. Thus, our findings reveal differences on thequaternary structure level. Lastly, these observationswill be helpful for future efforts to obtain high-resolu-tion information on the structure of BSE prions.

21. Proximity-dependent biotinylation identifiesprotein interactors of pathological tau andsynuclein in human ESC-derived neurons

Tagan A. Griffin, Elisa M. Cleveland, Robert W.Newberry, George A. Carlson, Paul D. Schnier andStanley B. Prusiner

Institute for Neurodegenerative Diseases, Weill Institute forNeurosciences, University of California, San Francisco, CA, USA

CONTACT Tagan A. Griffin [email protected]

ABSTRACT

Tau and synuclein can act as prions, undergoing templatedconformational changes resulting in the accumulation ofaggregated proteins, which drives the spread of neurode-generation throughout the brain. It is unclear which othercellular proteins contribute to this process. To investigatethis question, we utilized a promiscuous biotin ligase(BioID2) fused to tau or synuclein to biotinylate interactingproteins in human embryonic stem cell (ESC)–derivedneurons following treatment with disease-causing recom-binant fibrils (tau K18 or synuclein preformed fibrils[PFFs]). To identify interactors in a relevant cell type, wefirst generated a stable human ESC line (H1 ESCs) expres-sing neurogenin 2 (NGN2) under the control of an indu-cible promoter using PiggyBac transposon vectors. Upondoxycycline induction, this ESC line differentiated into anearly pure population of glutamatergic neurons in under3 weeks. Additional vectors were used to generate ESClines that differentiated into neurons expressing tau-BioID2 (WT or P301L), synuclein-BioID2 (WT orA53T), and a control line expressing BioID2 alone.Following neuronal differentiation, recombinant fibrilswere added to the culture media to induce aggregation,and cells were harvested 24 h or 3 weeks later. Following

8 ABSTRACT

denaturing lysis and streptavidin purification, the biotiny-lated proteins were subjected to quantitative mass spectro-metry using tandem-mass tag (TMT 10-plex) labels,enabling direct comparisons between multiple conditions(WT vs. mutant protein, ± fibrils, and early vs. late timepoints). We identified a number of known tau and synu-clein interactors as well as previously unreported candidateinteractors, many of which were less abundantly biotiny-lated in neurons treated with fibrils relative to neurons nottreated with fibrils. This was accompanied by a strikingincrease in the abundance of biotinylated tau or synuclein,consistent with aggregate formation and a concomitantloss of physiological interactions. We also found severalother biotinylated proteins whose abundances increasefollowing the addition of fibrils. Tau-BioID2 and synu-clein-BioID2 shared few interactors in common, consistentwith amodel of prion replication driven by tau or synucleinalone. These results identify novel candidate interactors oftau and synuclein, and shed light on the mechanisms ofprion infection and replication in human neurons.

22. Incapability of mutant PrPSc to recruit wild-type PrPC in a familial prion disease linked toPrPQ227X mutation implies a role of GPI anchor inprion propagation

Pingping Shena,b, Johnny Danga, Zerui Wanga,b, JueYuana, Yue Langa,b, Jiachun Fengb, QingzhongKonga,b,c, Annemiek J. M. Rozemullerd, Li Cuib,Robert B. Petersena,b, Wen-Quan Zoua,b,e

aDepartment of Pathology and Neurology, Case Western ReserveUniversity, Cleveland, OH, USA; bDepartment of Neurology, FirstHospital of Jilin University, Changchun, Jilin Province, China;cNational Prion Disease Pathology Surveillance Center, CaseWestern Reserve University, Cleveland, OH, USA; dDutchSurveillance Center for Prion Diseases, University Medical CenterUtrecht Heidelberglaan 100, Utrecht, and Netherlands Brain Bank,Amsterdam, The Netherlands; eFoundation of Science, CentralMichigan University College of Medicine, Mount Pleasant, MI, USA

CONTACT Wen-Quan Zou [email protected]

ABSTRACTCellular prion protein (PrPC) is a glycoprotein anchored tothe cell surface by a glycosylphosphatidylinositol (GPI)anchor. The key molecular event in the pathogenesis ofprion disease is the conversion of PrPC into its pathologicalisoform PrPSc. Several lines of evidence have indicated thatthis event takes place on the cell surface and alternation ofPrPC localization on the cell surface by mediating the GPIanchor dramatically affects the formation and infectivity ofPrPSc. Notably, anchorless PrP in transgenic (Tg) miceproduced deposition of PrP amyloid in the brain, butcaused no neurological signs in animals. A patient with ananchorless PrPQ227X stop codon mutation was reported to

exhibit a GSS-like prion disease phenotype. Here we exam-ined the anchorless PrP from the brain of the patient withPrPQ227X mutation and compared the physicochemicalproperties of secreted and un-secreted PrP isoforms froma transfected human neuron line expressing PrPQ227X.Epitope mapping revealed for the first time that the PrPSc

from the patient with Q227X was derived from the mutantallele only. Fractionation of culturedhumanneuroblastomacells expressing human wild-type or Q227X mutant PrPdemonstratedmost wild-type PrP (PrPWT) being recoveredin the cell lysate fraction, andmost mutant PrPQ227X recov-ered in the medium fraction, suggesting that PrPWT wasattached to the cell surface, while PrPQ227X was secretedinto the media. In addition, the mutant PrP lacked the N-linked glycans found in PrPWT. Two small fragmentsmigrating at approximately ~6–8 kDa were identified inthe culture media, but not in cell lysate fraction. Finally,typical PrPSc three bands were amplified with normalhumanized transgenic mouse brain as the PrPC substrateby serial protein misfolding cyclic amplification. Our find-ing that PrPSc derived from themutant GPI-anchorless PrPfailed to recruit cell surface-attached PrPWT suggests thatthe GPI anchor plays an important role in prion propaga-tion when mutant and wild-type alleles are present in vivo.

Funding

Supported in part by the CJD Foundation and the NationalInstitutes of Health (NIH) NS062787 and NS087588 to W.Q.Z., NS062787 and NS109532 to W.Q.Z., and Q.K.,NS088604 to Q.K. as well as the National Natural ScienceFoundation of China [No. 81,671,186] to LC.

23. Inferring the effects of deer population andlandscape structure on spread of the chronicwasting disease in Alberta

Peter Smolkoa, Dana Seidelb, Evelyn Merrilla, MargoPybusa,c, Anne Hubbsc and Mark Ballc

aDepartment of Biological Sciences, University of Alberta,Edmonton, AB, Canada; bDepartment of Environmental Science,Policy & Management, University of California Berkeley, CA, USA;cAlberta Fish & Wildlife Division, Government of Alberta, Edmonton,AB, Canada

CONTACT Peter Smolko [email protected]

ABSTRACT

Chronic wasting disease (CWD) is a 100% fatal priondisease of cervids that has been detected in wild deer andelk and moose in two Canadian provinces. Alberta hashad a surveillance programme for CWD since 1996.CWD in wild deer in Canada was detected in adjacentSaskatchewan (2000) and later in Alberta (2005). Based

PRION 9

on data collected during the Alberta surveillance pro-gramme we assessed the risk of a harvested mule deerand white-tailed deer having CWD over a 15-year period(2000–2016) in eastern Alberta. We hypothesized thatthe probability of a hunter harvested deer being CWDpositive was a function of time since the disease was firstdetected in the area (2000), sex and age of the deer andcharacteristics of the area where the deer was harvested(agricultural areas, extent of woody cover, distance torivers and streams, topography, road density or distanceto human settlements, and proximity to other positiveCWD cases). We used previously developed risk modelusing surveillance data from 2000–2012 (n = 162 CWDcases) to predict risk in deer harvested in 2013–2016. Wethen redeveloped the risk model using data from 2000–2016 (n = 591) and tested the predictions of this modelusing positives from 2017 (n = 327). We also assessedhow species, sex, importance of site characteristics, anddistance to known CWD cases changed over time ininfluencing predicted risk of a deer having CWD in thecore area and on the edge of the CWD surveillance zone.The top ranked model supported a pattern of increasingprevalence of CWD in hunter harvested deer over time.The risk among mule deer was higher compared towhite-tailed deer and amongmales compared to females;however, the difference between species decreased andbetween sexes increased compared to early period. TheCWD risk was greater in areas close to rivers withabundant woody cover where surveillance and controlefforts should be focused.

KEYWORDS: Mule deer; white-tailed deer; transmis-sible spongiform encephalopathy; prion-protein; preva-lence; disease management

24. Effect of the exosomal prion protein on theamyloid-β aggregation kinetics

Mohsin Shafiqa*, Alexander Hartmanna*, Stefano DaVelab*, Christiane Mutha, Behnam Mohammadia, LuiseLinsenmeiera, Hermann Altmeppena, SusanneKrasemanna, Dimitri Svergunb and Markus Glatzela

aInstitute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; bEMBL European MolecularBiology Laboratory, Hamburg, Germany

CONTACT Mohsin Shafiq [email protected]

*These authors contributed equally to this work.

ABSTRACT

The cellular prion protein (PrPC), a neuronally enrichedmembrane protein, is shown to impart a dual role in thepathological progression of Alzheimer’s disease. PrPC

molecules present on the cell surface serve as receptorsfor Aβ oligomers (Aβo) and initiate neurotoxic signallingevents; in contrast, extracellularly shed and cleaved formsof PrPC along with exosomally expressed PrPC are sug-gested to accelerate the Aβ42 fibril formation by seques-tering the Aβo thereby playing a neuroprotective role.

Aim of the current study is to investigate the influ-ence of PrPC expressing (WT) and PrPC deficient (KO)exosomes on the Aβ42 oligomerization kinetics, utiliz-ing small angle X-ray scattering (SAXS).

Experiments were designed for measuring the SAXSscattering profiles of various structural intermediates ofAβ42 along the different time points in aggregationcascade, in the presence of PrPC-WT and PrPC-KOexosomes.

Preliminary modelling of our data shows that thepresence of both exosome variants (PrPC-KO and WT)leads to accelerated Aβ42 fibrillation. However, rela-tively higher levels of unbound in-solution Aβo werenoticed in the presence of PrP-KO exosomes, indicat-ing a decline in Aβ42 binding ability of PrPC-KO exo-somes compared to that of PrPC-WT exosomes. Weascribe from our data that the presence of the PrPC iscrucial for the Aβ-sequestering activity in exosomes.

25. Aqueous extraction of formalin-fixed paraffin-embedded tissue and detection of prion diseaseusing real-time quaking-induced conversion

Eric M. Nicholson, Trudy Tatum and Soyoun Hwang

United States Department of Agriculture, Agricultural ResearchService, National Animal Disease Center, Ames, Iowa, USA

CONTACT Eric M. Nicholson [email protected]

ABSTRACT

Formalin-fixed paraffin-embedded tissue (FFPET) ismainstay in the diagnosis of prion disease by immuno-histochemistry (IHC), the standard by which all otherTSE diagnostic protocols are judged. IHC offers advan-tages over diagnostic approaches that typically utilizefresh or frozen tissue with regard to sample preservation.However, IHC and the formalin-fixed tissues utilized forIHC do not typically couple well to amplification basedapproaches of prion disease detection. This limitation hasthe potential to hamper sensitivity of techniques compa-tible with FFPET as advances in amplification based tech-niques such as real-time quaking induced conversion(RT-QuIC) continue to enhance sensitivity. In thisstudy, we apply an approach based on previous workutilizing FFPET and an aqueous extraction step for wes-tern blot and ELISA based detection of prion disease.FFPET from prion disease transmission studies of scrapie

10 ABSTRACT

in sheep, chronic wasting disease in white-tailed deer ortransmissible mink encephalopathy in cattle were cut at5 µm thickness. Samples containing the tissue equivalentof as little as one 5 µm section can be used to readilydiscriminate positive from negative samples using RT-QuIC. The detection approach is avoiding the potentialof a mixed chemical/biological waste stream that wouldoccur if organic solvents were used in the removal ofparaffin from the same, and offers the diagnostic andresearch community an addition in the diagnosis of TSEs.

26. Exploring the structure of mouse prion fibrilsby site-directed spin-labelling and ESR spectroscopy

Kuan Yu Chua,b, Chien Lun Hungc, Yun-Wei Chiangd,Sunney I. Chana,e and Rita P.-Y. Chenb,d

aDepartment of Chemistry, College of Science, Nation TaiwanUniversity, Taipei, Taiwan; bInstitute of Biological Chemistry,Academia Sinica, Taipei, Taiwan; cDepartment of Chemistry, NationTsing Hua University, Taipei, Taiwan; dInstitute of BiochemicalSciences, National Taiwan University, Taipei, Taiwan; eInstitute ofChemistry, Academia Sinica, Taipei, Taiwan

CONTACT Kuan Yu Chu [email protected]

ABSTRACT

Introduction: Recombinant prion protein (PrP) is a 23kDa soluble protein without GPI anchor and N-glyco-sylation. The structure of the N-terminal domain isdisordered and the structure of the C-terminal domaincontains three α-helices, two β-sheets and one disul-phide bond connecting helix 2 and 3. To study thestructural conversion mechanism of full-length mouseprion protein (mPrP), we explore which segments ofthe protein are involved in this conformational transi-tion thus form the cross-β structure.Methods: Structure conversion of mPrP was studiedusing Electron Spin Resonance Spectroscopy (ESR)and Site-Directed Spin Labelling (SDSL). Two intrinsicCys of mPrP were mutated to Ala. At the selectedpositions, the residue(s) were replaced by Cys andlabelled with MTSSL. The spin-labelled PrPs wereseeded by WT PrP fibrils to form various spin-labelledfibrils. The spin-diluted fibrils were prepared by mixingthe spin-labelled PrP with WT PrP.Results: ESR results of N174R1/N181R1, N181R1/Q186R1, and T193R1 fibrils showed that the wholehelix 2 was converted into several β-strands and actedas part of the amyloid core by forming cross-β struc-ture. For helix 3, not the whole helix 3 is transformedinto cross-β structure during fibril formation. The localregion around residue 212 might maintain its helicityand connect two intermolecular β-sheets. The 217–224

region formed a β-strand and are involved in the cross-β structureConclusions: Part of helix 1 of mPrP was unfoldedduring fibril formation. However, helix 2 and part ofhelix 3 of mPrP were involved in conformational con-version and formed the cross-β structure. The N-term-inal part of helix 3 remained its helical structure.

27. Biodegradation of BSE prions in compost

Tim A. McAllistera, Shanwei Xub, Sujeema Abeysekaraa,c,Sandor Dudasc, Gordon Mitchelld and Stefanie Czubc

aAgriculture and Agri-Food Canada, Lethbridge Research andDevelopment Centre, Lethbridge, Alberta, Canada; bAlbertaAgriculture and Forestry, Lethbridge, Alberta, Canada; cCanadian andOIE Reference Laboratories for BSE, Canadian Food Inspection AgencyLethbridge Laboratory, Lethbridge, Alberta, Canada; dNational and OIEReference Laboratory for Scrapie and CWD, Canadian Food InspectionAgency, Ottawa, Ontario, Canada

CONTACT Tim A. McAllister [email protected]

ABSTRACT

To reduce the transmission of PrPBSE, specified risk mate-rial (SRM) that potentially contain PrPBSE is required to beremoved from the food and feed chains and disposed of viarendering and landfilling or incineration in NorthAmerica. However, composting could be a more costeffective means of disposing of SRM and be more suitablein areas where SRM pickup services or suitable landfills areunavailable. Our study investigated the degradation ofPrPBSE (classical BSE) in lab-scale composters over28 days and in stimulated field-scale composters over106 days using protein misfolding cyclic amplification(PMCA). Laboratory-scale composting was conductedusing 45 kg of feedlot manure with and without feathers.Compost was mixed at day 14 to generate a second heatingcycle, with temperatures ≥55°C occurred for 2 days in thefirst cycle and less than 1 day in the second cycle. Based onPMCA, PrPBSE was reduced by 1 log in compost with andwithout feathers after 14 days. After 28 days, PrPBSE wasreduced by 3 logs and between 3–4 logs in compost withand without feathers, respectively. For stimulated field-scale composting, compost piles were constructed using2,200 kg feedlot manure and mixed at days 44 and 78 togenerate three heating cycles. Temperatures ≥55°C wereachieved for 67 out of 106 days of composting. However,after 106 days composting reduced PrPBSE by only 1–2 logs.Our findings showed that enrichment for proteolyticmicroorganisms through the addition of feather keratinto compost enhances the degradation of PrPBSE.However, other factors such as varying concentrationsand strains of PrPBSE used and nature of proteolytic micro-bial populations in compost may cause differential results

PRION 11

in the degradation of PrPBSE in lab and field-scale compost-ing systems.

28. Optimization of RT-QuIC for detection ofseeding activity in preclinical blood samples fromprion-infected sheep

Charlotte M. Thomas, Sandra McCutcheon, Richard A.Blanco, Boon Chin Tan and Fiona Houston

The Roslin Institute, R(D)SVS University of Edinburgh, Easter Bush,Edinburgh, Scotland, UK

CONTACT Charlotte M. Thomas [email protected]

ABSTRACT

Recent large-scale surveys of appendix samples estimatethat as many as 1 in 2000 people in the UK haveabnormal accumulation of disease-associated PrP, sug-gesting that they may be subclinically infected withvariant Creutzfeldt-Jakob disease (vCJD) [1]. If theseindividuals are infectious, they pose a potential risk toothers through blood and organ donation, or contam-ination of surgical instruments. Indeed, several cases ofvCJD have been attributed to receiving blood productsfrom individuals who donated blood during the pre-clinical stage of the disease [2]. Thus, a robust diagnos-tic test for vCJD is urgently required to detect these‘silent’ (preclinical/subclinical) infections, in order toprotect the UK blood supply.

Efforts to develop such a test are complicated by theextremely low concentrations of PrPSc present in readilyaccessible biological samples, such as blood, and this valueis likely to be even lower in preclinical samples. Recently,several platforms have been developed which may besensitive enough to overcome this challenge. One suchtechnique, an ultrasensitive in vitro prion amplificationassay, termed ‘real-time quaking induced conversion’(RT-QuIC), has already proven to be a highly effectivetool in the diagnosis of sporadic Creutzfeldt-Jakob disease(sCJD), when applied to samples of cerebrospinal fluid.[3] However, attempts to develop RT-QuIC into a blood-based diagnostic test for vCJD have been less successful.

Here we present a novel version of the RT-QuICreaction, using a previously untested recombinantprion protein (recPrP) substrate. Due to a paucity ofsuitable human samples (which are necessary to opti-mize the platform and evaluate its performance indetecting preclinical infection), we have optimized ourassay using an animal model of vCJD infection – sheepthat have been experimentally infected with BSE. Weshow that our version of the RT-QuIC assay givespositive amplification results when applied to wholeblood samples spiked with BSE-infected sheep brain

homogenate. Positive samples are amplified within20–40 h, with a high degree of analytical sensitivity,equivalent to a 10−6 dilution of BSE-infected brainhomogenate. We then assess the performance of theassay on ‘endogenously’ infected whole blood samples,collected from BSE-infected sheep at regular timepoints, from the time of infection to the onset ofclinical signs. If this version of the RT-QuIC reactiondemonstrates high levels of sensitivity and specificity inidentifying blood samples from pre-clinically infectedindividuals, it could then be adapted for use in humans.

References

[1] Gill O, Spencer Y, Richard-Loendt A, et al. Prevalentabnormal prion protein in human appendixes afterbovine spongiform encephalopathy epizootic: largescale survey. BMJ. 2013;347:f5675.

[2] Peden AH, Head MW, Ritchie DL, et al. PreclinicalvCJD after blood transfusion in a PRNP codon 129heterozygous patient. Lancet 2004;364:527–529.

[3] McGuire LI, Peden AH, Orrú CD, et al. Real timequaking-induced conversion analysis of cerebrospinalfluid in sporadic Creutzfeldt-Jakob disease. AnnNeurol. 2012;72(2):278–285.

29. High efficiency detection of all prion subtypesof sporadic Creutzfeldt-Jakob disease by PMCA

Adam Lyona, Sandra Pritzkowb, Silvio Notarib, BrianApplebyb, Pierluigi Gambettib and Claudio Sotoa

aFrom the Mitchell Center for Alzheimer’s Disease and Related BrainDisorders, Department of Neurology, University of Texas HoustonMedical School, Houston, Texas, USA; bNational Prion DiseasePathology Surveillance Center, Case Western University School ofMedicine, Cleveland, OH, USA

ABSTRACT

Sporadic Creutzfeldt-Jakob disease (sCJD) is a fatal neu-rodegenerative disorder representing 85% of humanprion diseases. It occurs at a rate of approximately onecase per 1,000,000 people per year and its etiology isunknown. There are at least five subtypes of sCJD clas-sified by the methionine (M)/valine (V) polymorphismat codon 129 and by the electrophoretic mobility of theprotease-resistant core fragment of PrPSc. The unglyco-sylated band of PrPSc type 1 is ~21 kDa, and that ofPrPSc type 2 is ~19 kDa. The molecular classificationaligns with distinct clinical and pathological features.While all subtypes remain invariably fatal, early andaccurate diagnosis of sCJD and other human prion dis-eases is important for patients and their families, as wellas for health care providers. Detection by real-timequaking induced conversion (RT-QuIC) of sCJD prionsin CSF and from nasal brushings has demonstrated high

12 ABSTRACT

sensitivity and specificity, but the challenge remains todetect sCJD prions in fluids and tissues whose collectionis less invasive. Previously, we have shown successfuldetection of variant CJD prions in blood and urine byprotein misfolding cyclic amplification (PMCA). Themain goal of this study was to adapt the PMCA assayfor high sensitive detection of all forms of sCJD to allowtype identification of sCJD PrPSc in patients’ blood andurine. Our results demonstrate efficient amplification ofall sCJD subtypes by PMCA in brain homogenate, with avariable rate depending on the specific strain. Indeed, wecan reproducibly detect at least a 10−7–10−11 dilution ofbrain homogenate from all samples analysed. Currently,we are analysing blood and urine samples from patientswith sCJD with diverse genotypes. This is a crucial steptowards implementing PMCA as a general tool for rou-tine diagnosis of sCJD including sCJD subtype, and toscreen blood to increase the safety of the blood supply.

Funding

SB1NS079060, R01 NS083687, P01 AI106705 andCharles S. Britton Fund (PG).

30. A blue light optogenetic and CRISPR/Cas9system for inducible PrPC expression

Luis A. Arcea,b, Serene Wohlgemutha and DavidWestawaya,b

aCentre for Prions and Protein Folding Diseases; bDepartment ofBiochemistry, University of Alberta, Edmonton, Alberta, Canada

CONTACT Luis A. Arce [email protected]

ABSTRACT

An inducible system for expression of wild type PrPC canoffer advantages for investigating the chemical biology ofthis GPI-linked cell-surface protein. Previous induciblePrPC systems have utilized non-physiological xenobioticcompounds such as tetracycline to control gene expression[1,2]. However, wishing to avoid off-target chemical effectsof xenobiotic additives, we are developing an induciblesystem controlled by light. CRISPR/Cas9 systems that acti-vate transcription (CRISPRa) utilize transcriptional activa-tion domains (TAD), as found in VP64 or p65 proteins,attached to a modified Cas9 (called dCas9) [3]. dCas9 hasinactivated endonuclease function but can still be directedto a specific genomic locus by a ‘guide RNA’ (gRNA);previous studies have exploited this interaction by thedevelopment of dCas9/TAD fusion proteins that allow forthe activation and overexpression of targeted genes [4].Inducible CRISPR-based systems have been developedtypically using a split-Cas9 method and small effectormolecules, such as rapamycin. However, these systems

can also exhibit leakiness [5]. An alternative system whichutilizes the light inducible protein interactions betweencryptochrome 2 (CRY2) and CIB1 from Arabidopsis thali-ana has proven to be effective in both gene induction andmitigation of residual transcriptional activity [6,7]. Thesystem contains two fusion proteins; the ‘anchor’ (dCas9/CIB1), and the ‘activator’ (CRY2/TAD); targeting of dCas9is achieved by the gRNA and then heterodimerization ofCRY2 and CIB1 is based upon blue light irradiation. Herewe describe adaptation of this optogenetic system to mod-ify transcription initiated from the human Prnp promoterregion. Following a gRNA design phase, correspondingsequences were inserted into an expression vector. Serialtransfections were used to deliver the two fusion proteinsplasmids and designed gRNA plasmids into the humanHAP1 cell line. Transfected cells are subjected to blue lightirradiation and then assessed for PrPC levels using quanti-tative capillary westerns. This inducible system may offeradvantages for drug screening to identify novel chemicalsthat modify PrPC biology.

KEYWORDS: CRISPR Cas9; optogenetics; PrPC; drugscreen

References

[1] S. Huang, et al. PNAS 2007;104(16):6800–6805.[2] E. Sabuncu, et al. Biochem Biophys Res Commun

2005;337(3):791–798.[3] L. A. Gilbert, et al. Cell 2013;154(2):442–451.[4] M. E. Tanenbaum, et al. Cell 2014; 159(3):635–646[5] B. Zetsche, et al. Nat Biotechnol 2015;33(2):139–141.[6] M. J. Kennedy, et al. Nat Methods 2010;7(12):973–975.[7] Y. Nihongaki, et al. Chem Biol 2015;22(2):169–174.

31. Exploring the role of PrP in axonal tractformation during zebrafish nervous systemdevelopment

Fabrizio Cuevasa,b, Ignacio Gutierreza,b, Carmen G.Lemusa,b and Miguel L. Conchaa,b,c

aAnatomy and Developmental Biology, Institute of BiomedicalSciences, Faculty of Medicine, Universidad de Chile, Santiago,Chile; bBiomedical Neuroscience Institute, Santiago, Chile; cCenterfor Geroscience, Brain Health and Metabolism, Santiago, Chile

CONTACT Fabrizio Cuevas [email protected]

ABSTRACT

Although the role of PrPc in neurite outgrowth, neuronaldifferentiation and synapse formation has been describedin vitro, little is known of this phenomenon in vivo andespecially during development. Zebrafish (Danio rerio) hasemerged as a promising model to study PrP physiologicalfunction because of its external development, embryo

PRION 13

transparency and amenability to geneticmanipulation. Thedevelopmental expression of genes encoding for the zebra-fish PrP paralogs prp-1 and prp-2 is still controversial thuswe first revisited in detail the spatiotemporal expression ofboth genes by in situ hybridization. prp2 is expressed from18 h post fertilization (hpf) in the encephalon and the firstsegments of the spinal cord, and also in structures outsidethe nervous system. In contrast, prp1 is detectable in thecentral nervous system only from 48 hpf, and in cranialganglia from 60 hpf. These results are consistent with aprevious report [1], but differ in the lack of expression seenat earlier stages. To begin exploring the function of PrP inaxonal tract formation we perfomed acetylated tubulin byindirect immunofluorescence in mutant embryos and lar-vae lacking prp-1 and prp-2 function. We found that prp-1and prp-2 mutants both show weaker intensity of anti-acetylated tubulin staining and less axonal arborization insome specific brain regions, like the olfactory bulb andcranial ganglia, and thinner tracts at the level of the cere-bellar commissure. The presence of abnormal tracts in prp-1 and prp-2mutants is intriguing given the normal appear-ance of embryos and larvae.We are currently dissecting theaxonal tract deficiencies using fluorescent reporters in prp-1 and prp-2 mutants.

Funding

CONICYT PFCHA/DOCTORADO BECAS CHILE/2017–21,171,130, ICM P09-015-F and Fondap 15,150,012.

Reference

[1] Cotto E, Andre M, Forgue J, et al. Molecular character-ization, phylogenetic relationships, and developmentalexpression patterns of prion genes in zebrafish (Daniorerio). Febs J. 2005;272(2):500–13.

32. The EV-export pathway for misfolded proteins

Desmond Pinka, John Lewisa,b and Janice E.A. Braunc

aNanostics Precision Health, Edmonton, Alberta, Canada; bHotchkissBrain Institute, Department of Biochemistry and Molecular Biology,Cumming School of Medicine, University of Calgary, Calgary, Alberta,Canada; cDepartment of Oncology, University of Alberta, Edmonton,Alberta, Canada

CONTACT Janice E.A. Braun [email protected]

ABSTRACT

Extracellular vesicles (EVs) are a collection of secretedvesicles of diverse size and cargo that are implicated inthe physiological removal of nonfunctional proteins aswell as the cell-to-cell transmission of disease-causing-proteins in several neurodegenerative diseases. Wehave shown that the molecular chaperone, cysteine

string protein (CSPα; DnaJC5), is responsible for theexport of disease-causing-misfolded proteins fromneurons via EVs. We report here that CSPα-EVs effi-ciently deliver GFP-tagged 72Q huntingtinexon1 tonaive neurons. When we analysed the heterogeneousEV pool, we found that the misfolded GFP-tagged72Q huntingtinexon1 cargo was primarily found inEVs between 180–240nm. We further determinedthat cargo-loading of GFP-tagged 72Q huntingtinexon1

into EVs was impaired by resveratrol. In addition toCSPα, we identified two other J protein co-chaper-ones, DnaJB2 and DnaJB6 that facilitate EV exportof GFP-tagged 72Q huntingtinexon1. While humanmutations in CSPα cause the neurodegenerative dis-order, adult neuronal ceroid lipofuscinosis, mutationsin DnaJB6 cause limb-girdle muscular dystrophy andmutations in DnaJB2 are linked to the neurodegenera-tive disorders, Charcot Marie Tooth disease, distalhereditary motor neuropathy, spinal muscular atrophyand juvenile Parkinsonism. Our data provides newinsights into the parallels between proteostasis andEV export, as three J proteins linked to disease inhumans are the same as those that mediate EV genesisand export of misfolded proteins.

33. Detection of CWD prions in third eyelids ofdeer and elk

Sarah K. Coopera, Clare E. Hoovera,b, Davin M.Hendersona, Nathaniel D. Denkersa, Candace K.Mathiasona and Edward A. Hoovera

aPrion Research Center, Department of Microbiology, Immunology,and Pathology, College of Veterinary Medicine and BiomedicalSciences, Colorado State University, Fort Collins, CO, USA;bAstraZeneca, Waltham, NJ, USA

CONTACT Sarah K. Cooper [email protected]

ABSTRACT

Background: The increasing prevalence of CWDglobally,makes critical the development of fast, cost-effectivemethods to detect the disease in hunter-harvested deerand assist wildlife population disease management. Basedin part on the demonstration of PrPSc in the third eyelidlymphoid follicle in sheep with scrapie, we explored thethird eyelid for its potential as a non-invasive and easilyaccessible lymphoid tissue that could be sampled withoutspecial anatomical training antemortem and postmortemfor PrPCWD detection by real-time quaking induced con-version (RT-QuIC) and immunohistochemistry (IHC).Methods: We compared RT-QuIC detection sensitivity inthe third eyelid to the retropharyngeal lymph node andobex region of the brain from experimentally CWD inocu-lated white-tailed deer and naturally occurring subclinical

14 ABSTRACT

elk using IHC. We examined symptomatic and asympto-matic white-tailed deer inoculated with CWD(+) andCWD(-) brain homogenate or saliva via the per os or aero-sol route. In addition, we examined samples from asympto-matic, naturally exposed elk which were culled due to beingRAMALT biopsy positive or suspect by RT-QuIC.Results: We identified prion seeding activity in thirdeyelids in 24 out of 25 (96%) samples from terminal,experimentally CWD-infected deer and found RT-QuICpositivity in the third eyelid as early as 1 month afterexperimental exposure to CWD. In addition, we identi-fied prion seeding activity in third eyelids in 17 out of 25(68%) samples asymptomatic, naturally exposed elk. Bycontrast, IHC detected prion deposition in third eyelidlymphoid follicles in 5 of 10 deer (50%). We show thatRT-QuIC can be more sensitive for CWD prion detec-tion in the third eyelid compared to IHC.Conclusions: RT-QuIC testing on the third eyelid is arapid and sensitive means for post-mortem detection ofCWD with potential to augment CWD diagnostics andhunter testing compliance.

Funding

Supported by NIH R01-NS-061902, P01-AI-077774,F30-ODO-118,143, T32-OD0-10,437

34. microRNAs profiling in patient bloods identifiesa novel signature associated with sCJD diseaseprogression

Penny Norsworthy, John Collinge, Simon Mead andEmmanuelle Viré

MRC Prion Unit at UCL, Institute of Prion Diseases, Queen SquareHouse, Queen Square, University College London, London, UK

CONTACT Emmanuelle Viré [email protected]

ABSTRACT

Prion diseases are fatal neurodegenerative diseases ofhumans and animals caused by the misfolding andaggregation of prion protein (PrP). Misfolded proteinsplay an important role in many forms of dementia butdespite intensive efforts to better understand the patho-physiology of these diseases, the mechanisms involvedare still poorly understood. Although neurodegenerationis affecting millions of people worldwide there is a hugeunmet need for novel diagnostic and treatmentapproaches in neurodegenerative disorders to countertherapeutic nihilism in this area. Recent studies haveimplicated a role for genetic and epigenetic variation inneurodegenerative disorders. To date, however, no studyhas systematically explored the contribution of epige-netic changes in human prion diseases.

microRNAs (miRNAs) play crucial role in human dis-eases, and the discovery of the presence of cell-freemiRNAsin the blood stream probed researchers to investigate therole of miRNAs as potential non-invasive biomarkers.

We performed genome-wide microRNA deep sequen-cing on 107 blood samples frompatients with sporadic CJDand healthy controls and identified five differentiallyexpressed microRNAs. We confirmed that this miRNAsignature is specific to sporadic CJD since the level ofthose 5miRNA remained unchanged in other neurodegen-erative disorders (CJD and Alzheimer’s diseases).Investigating longitudinal samples from more than 20patients with sCJD, we found that the miRNA signaturestrongly correlates with disease severity. These results havebeen replicated using both an independent cohort andqPCR technology.

Integrating our data, we have established a sCJD spe-cific signature, with genes between the two groups beingenriched in functions related to neuroinflammation andimmunity; and have correlated it with age of onset, dis-ease progression, severity, decline, and genotype.

In light of our findings, we will discuss the potentialimplications for this novel gene signature to comple-ment existing non-invasive biomarkers and its rele-vance for disease management.

35. Process of structural diversification andsynergy between prion subassemblies during hostadaptation

Angélique Igel-Egalona, Mohammed Moudjoua,Florent Laferrièrea, Philippe Tixadora, Jan Bohla,Mathieu Mezachea,b, Tina Knäpplea, Laetitia Herzoga,Fabienne Reinea, Marie Doumicb, Vincent Béringuea

and Human Rezaeia

aVIM, INRA, Université Paris-Saclay, Jouy-en-Josas, France; bINRIA,MAMBA, Université Paris VI, Paris, France

CONTACT Human Rezaei [email protected]

ABSTRACT

In prion diseases, the cellular prion protein PrPCcan misfold into PrPSc and auto-organize into con-formationally distinct assemblies or strains. Aplethora of observations reports the existence ofPrPSc structural heterogeneity within prion strains,suggesting the emergence and coevolution of struc-turally distinct PrPSc subassemblies during prionreplication in controlled environment. However,the nature of and the underlying processes of thestructural diversification remain poorly exploredand mainly based on conjectural observations.Conceptually, such structural heterogeneity appears

PRION 15

central to prion host adaptation since the actualparadigm consists mostly on the best replicatorselection, i.e. selection of the best-fit subset ofPrPSc assemblies within the PrPSc structural cloud.

Here, we first characterized the evolution of the PrPScquaternary structure in early versus late stage of prionreplication (in a homotypic PrP context) in vivo and inbona fide cell-free amplification assays. Regardless of thestrain studied, the early replication stage conduced to thepreferential formation of small PrPSc oligomers, thushighlighting a quaternary structural convergence phe-nomenon. The evolution of these small oligomeric assem-blies led to the formation of structurally differentmultimers through a secondary templating pathway inan autocatalytic and PrPC-dependent manner. Second,we addressed the role of such structural PrPSc diversifica-tion in prion adaptation on cross-species transmission.We isolated by size-fractionation or by dilution theformed PrPSc assemblies and measured their relativepropensity to cross an existing species barrier, as mod-elled by transgenic mice expressing heterotypic PrPC. Weshowed impaired and aberrant fitness of the isolatedPrPSc subassemblies as compared to the unfractionatedand undiluted populations, respectively.

Altogether our observations uncover the existence oftwo mechanistic processes during prion replication:first, prion structural diversification occurs through asecondary templating pathway. Second, overcomingspecies barriers requires complementation between thePrPSc subassemblies. These processes explain howstructurally diverse assemblies can coexist despite com-petition for the same PrPC substrate and truly chal-lenge the best- replicator selection models.

36. Structural characterization of amyloidogenicproteins implicated in neurodegeneration underconditions of intracellular crowding and in non-polar environments

Nikolay Blinova,b, David S. Wishartc, Neil R. Cashmand,and Andriy Kovalenkoa,b

aUniversity of Alberta, Department of Mechanical Engineering,Edmonton, Canada; bNRC Nanotechnology Research Centre,Edmonton, Canada; cUniversity of Alberta, Departments of BiologicalSciences and Computing Science, Edmonton, Canada; dUniversity ofBritish Columbia, Department of Medicine, Vancouver, Canada

CONTACT Nikolay Blinov [email protected]

ABSTRACT

Misfolding and aggregation of proteins is a key patho-logical event in many neurodegenerative disorders,including the prion diseases, Alzheimer’s disease(AD), and Amyotrophic Lateral Sclerosis. Structural

properties of proteins and their aggregates (oligomersand amyloid fibrils) are essentially affected by theirsolvation environments, the presence of metabolitesand salt ions. Structural characterization of oligomerswhich may be the most toxic agents in neurodegenera-tion is a very important task because the inhibition offormation of these aggregates and their pathologicalaction has been proposed as a therapeutic target tofight neurodegeneration.

Experimental structural studies are complicated bythe transient nature of oligomers. In the case of AD,amyloid (A)β peptides (constituent units of the neuro-toxic oligomers responsible for neurotoxicity in AD) areintrinsically disordered, which introduces another levelof complexity to the problem. Despite the significantefforts, there is no consensus on structure and aggrega-tion pathways of Aβ peptides in cellular environments.The situation is further complicated by the fact that thepeptides can aggregate and perform their neurotoxicactivity intracellularly. The intracellular Aβ species canalso be secreted into the extracellular space where theymay trigger prion-like pathological misfolding.

In this poster, we provide an overview of our recentefforts towards structural characterization of the amyloido-genic proteins and their aggregates under different solventenvironments. The new methodology has been developedbased on the 3D-RISM-KH molecular theory of solvationto account for the effects of specific conditions of intra andextracellular environments on the conformational equli-brium of intrinsically disordered peptides. Applied to AD,it shows the similarities and differences between conforma-tional properties of Aβ peptides under conditions of intra-cellular crowding and in non-polar environments (such asimposed by the neuronal lipid membrane). Specifically,both the crowding and the non-polar solvation make thepeptides less disordered, which may result in the increaseof their propensity for aggregation. Among the differencesare the opposite trends in bend and turn secondary struc-ture contents under different conditions. We also showthat the 3D-RISM-KH based methodology could be usedto predict effects of synaptic metabolites and other smallmolecules (e.g. therapeutic agents) on the structure of thepeptides. This feature of the methodology can be used toscreen potential inhibitors of pathological misfolding ofproteins in neurodegeneration.

37. Spontaneous neurodegeneration in transgenicmice caused by histidine to tyrosine substitution atnon-OR copper-binding site

Alba Marín-Morenoa*, Thanh Hoa Tranb*, Juan CarlosEspinosaa, Edoardo Bistaffac, Fabio Modac, Juan MaríaTorresa and Giuseppe Legnameb

16 ABSTRACT

aCentro de Investigación en Sanidad Animal (CISA-INIA), Madrid,Spain; bLaboratory of Prion Biology, Department of Neuroscience,Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy;cFondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy

CONTACT Alba Marín-Moreno [email protected]

*These authors contributed equally to the work

ABSTRACT

Prion protein has long been known as a copper bindingprotein via histidine residues in the octapeptide repeats(OR) and the non-OR region located in the disordered N-terminal tail of the protein. The functional implication ofcopper binding to PrP is not yet clear, but copper seems toplay an important factor in prion disease.

Transgenic mice expressing the mouse prion proteinreplacing histidine (H) by tyrosine (Y) at codon 95 todisrupt the non-OR copper-binding site have beengenerated.

Mice overexpressing approximately two fold PrP H95Yshowed clinical signs and died at about 100 days withspongiform degeneration and atypical PK-resistant PrP inthe brain. Inoculation of brain homogenate from termin-ally ill mice overexpressing PrP H95Y to mice expressing alow level of PrP H95Y (1 fold) or Tga20 mice also causeslethal, spongiform encephalopathy.

H95Y substitution at non-OR region could promotePrPC-PrPSc conversion and induce spontaneous neurode-generation disease in transgenic mice, implying a criticalrole for the non-OR copper binding site in prion disease.

KEYWORDSScrapie; prion strain; transmission barrier; mouse bioassay

Funding

Spanish Ministerio de Ciencia, Innovación yUniversidades (AGL2016-78,054R).

38. Signalling pathways underlying prionneurotoxicity

Nhat T. Le, Robert C. Mercer and David A. Harris

Department of Biochemistry, Boston University School of Medicine,Boston MA, United States of America

ABSTRACT

The process by which prions propagate is nowwell under-stood; however, the molecular mechanism responsible fortheir neurotoxicity remains unclear. Synaptic loss is oneof the earliest events in both in vivo and in vitromodels ofprion disease. Recently, we have developed sophisticatedneuronal cell culture models to analyse the mechanisms

of prion-induced synaptic degeneration in a more phy-siologically relevant setting. Using this system to dissectthe underlying cellular and molecular mechanisms, weshow that exposure of hippocampal neurons to PrPSc

results in rapid, PrPC-dependent retraction of dendriticspines. In this process, PrPSc activation of AMPA andNMDA receptors induces calcium influx, which subse-quently activates p38 MAPK and its downstream MK2/3kinases. Pharmacologically and genetically reducing p38MAPK activity not only blocks PrPSc-induced loss ofdendritic spines, but also reverses dendritic spine lossthat has already occurred. In order to identify additionalcomponents of prion-induced synaptotoxic pathways, wemeasured changes in the phosphoproteome and tran-scriptome of cultured neurons exposed to PrPSc whilethey are undergoing well-defined alterations in synapticstructure and function. This work identifies a new, drug-gable target that could be used to block synaptotoxicsequelae in prion diseases: p38 MAPK. We are in theprocess of testing whether reducing p38 MAPK function,either genetically or pharmacologically, ameliorates thesymptoms of prion disease in mice.

39. Generation of bona fide prions by large-scaleProtein Misfolding Cyclic Amplification

Fei Wang, Luis Concha-Marambio, Enrique Armijoand Claudio Soto

Mitchell Center for Alzheimer’s Disease and Related Brain Disorders,Department of Neurology, McGovern Medical School, University ofTexas Health Science Center at Houston, Houston, Texas, USA

CONTACT Fei Wang [email protected]

ABSTRACT

The misfolding of normal prion protein (PrPC) intopathogenic PrPSc is the key event causing prion dis-eases. However, the molecular mechanism underlyingsuch conformational conversion remains elusive, lar-gely due to the absence of high-resolution structuralinformation of PrPSc, hence impeding the develop-ments of efficacious therapeutic strategies againstthese currently incurable diseases. To facilitate thestructural studies of PrPSc, here we present our latestresults of large-scale production of bona fide prions byProtein Misfolding Cyclic Amplification (PMCA),which not only provides the platform for ultra-sensitivedetection of prions in biological samples, but can alsofaithfully reproduce the key properties of prions,including infectivity, species barrier, and strain varia-bility. In a pilot study, normal mouse brain homoge-nates were seeded with RML prion in large containersto produce bona fide prions by PMCA in reaction

PRION 17

volumes ranging from 30 mL to 120 mL. The prionyielded in this large-scale production maintained theRML prion strain characteristics in terms of infectivityin wild type animals, incubation time, PrPSc biochem-ical profile and pathological changes. In the follow-upstudy, recombinant prion generated from bacteriallyexpressed full-length recombinant mouse PrP seededwith RML prion was used to seed the large-scale pro-duction of recombinant PrPSc by PMCA. The large-scale PMCA (LS-PMCA) platform is capable of produ-cing at least 2 mg of PK-resistant recombinant PrPSc ina 200-mL reaction in 24 h and has the option to scaleup to as much as 1000 mL per round of LS-PMCA. Theinfectivity assessment of the recombinant PrPSc fromLS-PMCA is currently ongoing. Availability of a proce-dure to produce massive amounts of PrPSc usingrecombinant protein as substrate which maintain thetypical biological and biochemical properties of bonafide prions, will facilitate structural and biochemicalstudies of infectious prions.

40. Impact of different recombinant PrP substrateson the signalling response in the RT-QuIC

Susana Correia, Matthias Schmitz and Inga Zerr

Department of Neurology, Medical University of Goettingen,Goettingen, Germany

CONTACT Susana Correia [email protected]

ABSTRACT

Transmissible spongiform encephalopathies (TSE) are agroup of neurodegenerative disorders, such asCreutzfeldt-Jakob disease (CJD), which are based onthe conformational change of the prion protein (PrP)from the cellular form, PrPC, to the infectious scrapieform, PrPSc.

Based on the mechanism that PrPSc acts as a tem-plate for the conversion of PrPC, protein misfoldingamplification assays, such as the real-time quaking-induced conversion (RT-QuIC) has been applied forthe detection of misfolded PrPSc in the brain andcerebrospinal fluid (CSF) for the pre-mortem diagnosisof human prion diseases.

The technique bears a huge diagnostic and analyticalpotential. However the clinical variability of differentsCJD subtypes (MM1, MM2, MV1, MV2, VV1, andVV2) has a relevant impact on the accuracy of diagnosticassays. An additional important factor is the kind ofrecombinant PrP substrate (e.g. hamster, hamster-sheepchimera, bank vole, etc.) used in the RT-QuIC. The diag-nostic accuracy for different sCJD subtypes, familialforms – or the variant form of CJD is variable, e.g. only

a very few recombinant PrP substrates are able to detectvariant CJD prions, such as bank vole PrP.

In our study, we aim to increase the diagnosticaccuracy and the application spectrum of the RT-QuIC for all types of prion diseases, in particular forthe atypical and rare forms like sCJD (MM2T, MM2C,and VV1), FFI or GSS. We will produce and purify apanel of different recombinant PrP substrates and com-pared the sensitivity and specificity among them acrossthe wide spectrum of prion diseases to reach an opti-mized diagnostic accuracy.

41. A screen for prion and α -synuclein aggregatedecontaminants to enhance safety of surgicalinstrument reprocessing

Daniel Heinzera, Merve Avara, Manuela Pfammattera, RitaMoosa, Linda Irpinioa, Dezirae Schneidera, BenjaminKuhnb, Stefan Mauerhoferb, Urs Rosenbergb, AdrianoAguzzia, Simone Hornemanna

aInstitute of Neuropathology, University of Zurich, Zurich,Switzerland; bBorer Chemie AG, Zuchwil, Switzerland

CONTACT Simone Hornemann [email protected]

ABSTRACT

Prions, the infectious agents causing prion diseases suchas the Creutzfeldt-Jakob disease, show extreme robustnessto common decontamination procedures. Therefore,there is a risk of iatrogenic transmission of these lethaldiseases by inefficiently decontaminated medical instru-ments. So far, improvement of effective decontaminatingagents applicable to routine sterilization procedures hasnot been feasible as the traditional assays for measuringprion infectivity are time-consuming and constrained bya limited throughput. The development of the real-timequaking induced conversion (RT-QuIC) assay [1], a fastand ultra-sensitive method, based on the self-propagationof proteinaceous agents in misfolding diseases, provides anew approach for the detection of infectious prions.

The adaption of this assay to a carrier mimickingsurgical steel allows evaluating the efficiency of novelpotential decontaminants in a fast and proficient way. Inaddition, with the emergence of evidence of α-synuclei-nopathies exhibiting prion-like infectious properties, thismethod was further adapted and applied to assess inacti-vation procedures for potentially hazardous α-synucleinfibrils. We demonstrated that the prion RT-QuIC assay issuitable to detect propagating prions adsorbed to 316Lsteel type metal beads, and that the treatment with thestandard prion decontamination procedure (1 M NaOHfor 2 h) completely abolished a positive signal in the assay.Around 120 different formulations were tested for their

18 ABSTRACT

efficiency to inactivate or remove prions from the steelbeads. We identified one potential candidate formulationwith good material compatibility that strongly and reli-ably reduced prion propagation activity in the RT-QuICassay. The candidate formulation and two further alkalinecleaner are currently validated in a cell and mouse bioas-say to confirm their decontamination properties. In addi-tion, we developed an α-synuclein RT-QuIC assay that weused to test the inactivation efficiencies of known priondecontaminants on α-synuclein preformed fibrils.Interestingly, some of the commonly used prion deconta-mination procedures were less effective to deactivate thepropagation efficiency of α -synuclein preformed fibrils.This implies that the resistance of α -synuclein fibrilsmight differ or even exceed the one of bona fide prions,and that other decontamination procedures need to beapplied for the effective deactivation of α-synuclein fibrils.

Reference

[1] Wilham JM, Orrú CD, Bessen RA, et al. Rapid end-point quantitation of prion seeding activity with sensi-tivity comparable to bioassays. PLoS Pathog. 2December 2010;6(12):e1001217.

42. Multiple sclerosis brain transmits pathology tohumanized transgenic mice

Shigeki Tsutsuia, Shoraf Dadakhujaeva, FranciscaCavalcante Meloa, Geert Schenkb, Roel Klaverb,Hugo Tedforda, Najwa E Kadia, Karen Cumminsa,Gui Fang Guoa, Andrew V Caprarielloa, JulianneProfta, Shyamosree Roychoudhurya, Kyoichi Tsutsuia,V. Wee Yonga, Jeffrey T Josepha, Gerald W.Zamponia, Jeroen JG Geurtsb and Peter K Stysa

aUniversity of Calgary, Calgary, AB, Canada; bVU University MedicalCenter, Amsterdam, Netherlands

ABSTRACT

The fundamental aetiology of multiple sclerosis (MS)remains unknown. Two competing theories proposethat on the one hand, MS is driven by a dysregulationof the peripheral immune system promoting inflamma-tory attacks on the CNS, while an alternative modelposits that an initial primary degenerative process maysecondarily trigger autoimmunity. Our hypothesis isthat MS, like many neurodegenerative diseases, mightbe a protein misfolding disorder driven by accumula-tion of pathological cytotoxic aggregates that canspread through the CSF and CNS. One defining prop-erty of such diseases is the transmission of pathology tosusceptible hosts.

We have previously shown that intracerebral inocula-tion (i.c.) of MS brain homogenates into human prionprotein (PrP) over-expressor mice induced significantMS-like pathology, together with cognitive disability.We have expanded our dataset using i.c. transmission ofMS pathologies in larger numbers of animals (53 micefrom 10 primary or secondary-progressive MS patients,65 mice from 9 control donors, 30 mice from 3 chronicencephalitis brains, and 10 mice from other neurologicaldiseases, such as AD and Lewy body disease), using addi-tional immunohistochemical injury markers, whichfurther supports the underlying hypothesis. Passaging(inoculation of naïve mice with brain homogenate ofMS-inoculated mice) also continued to transmit pathol-ogy. MS-injected mouse brains exhibited significantdegeneration of myelin and axonal damage with micro-glial and astroglial activation in the corpus callosum,leukocortical junction, thalamus and peri-ventricularregion, assessed by SMI-94, QD9, SMI-32, Iba-1 andGFAP immunohistochemistry, whereas mice injectedwith control human brain did not develop pathology.Importantly, PrP-immunodepleted MS brain homoge-nates, blunted transmission of pathology, further support-ing a possible role of pathological PrP. Formic acid-resistant prion protein aggregates were detected in bothwhite and grey matter regions. Protease resistance, oftenseen in more conventional prionopathies, was notdetected in either human MS brain or MS-inoculatedmice.

Conclusion: Our results are consistent with thehypothesis that MS might be a primary degenerativedisorder caused by accumulation and propagation ofatypical pathogenic PrP, that can transmit pathology tohumanized murine hosts. We speculate that these toxicpathological conformers could circulate in the CSF andspread to various brain regions in contact with CSF, as ischaracteristic of human MS lesion distribution. Theresulting degeneration of myelin and release of antigenicdebris could secondarily trigger an autoimmune inflam-matory response in immune-predisposed hosts.Together, such a convolution of a primary prion-depen-dent degeneration, with secondary inflammation couldexplain the broad spectrum of human MS phenotypes.

43. Molecular characterization of prion diseases inDenmark: the national reference center experiencefrom 2002 to 2018

Aušrinė Areškevičiūtėa, Helle Broholma, Linea C.Melchiora, Piero Parchib, Anna Bartoletti-Stellab, EvaL. Lunda

PRION 19

aDanish Reference Center for Prion Diseases, Department of Pathology,Copenhagen University Hospital - Rigshospitalet, Copenhagen,Denmark; bIRCCS Istituto delle Scienze Neurologiche di Bologna,Ospedale Bellaria, Bologna, Italy

CONTACT Aušrinė Areškevičiūtė [email protected]

ABSTRACT

We present a Danish cohort of prion disease cases withwell-characterized fresh frozen brain samples that pro-vides a great foundation for future research.

The aim of this study was an up-to-date molecularcharacterization and classification of all fresh frozen priondisease samples delivered to the Danish Reference Centerfor Prion Diseases in the period of 2002 and 2018.

Molecular characterization and classification of thesesamples was based on an improved immunoblottingmethod and sequencing of the whole prion proteingene coding region. Immuno- and histopathologicalfindings in the brain tissues were re-evaluates in severalcases. Available medical records were gathered for esti-mation of patients’ age at disease onset and death,disease duration, and disease clinical presentation.

Thirty-seven of forty-one samples were classified assporadic prion diseases with the following subtypes:MM/MV1 (n = 23), VV2 (n = 7), MM/MV1+2 (n =3), MV2 (n = 2), MM2 (n = 2). A case of CJD withsporadic phenotype that occurred to a mother and wifeof P102L mutation carriers was also a part of theDanish prion diseases cohort [1].

Four of forty-one cohort samples were found toharbor mutations in the prion protein gene. Two ofthose mutations were unique 5 and 8 octapeptiderepeat insertions [2]. The other two mutations foundin the cohort were a well-known P102L, and a novelT201S point mutations. The pathogenicity of T201Swas recently investigated and described [3].

Re-evaluation of the cohort samples revealed thatinitial disease classification was inaccurate or missingin nearly every 5th case, which is a strong argumentfor consideration to re-evaluate older samples astechniques with higher sensitivity and specificity arebeing developed, and the classification systemupdated.

References

[1] Areškevičiūtė, et al. JNEN 2018;77:673–684.[2] Areškevičiūtė, et al. A novel eight octapeptide repeat

insertion in PRNP causing prion disease in a Danishfamily. JNEN;2019:In Press.

[3] Mok, et al. Neurobiol Aging. 2018;71:265.e1–265.e7.

44. Understanding prion-like mechanisms intraumatic brain injury using a novel in vivo model

Hadeel Alyenbaawia,b and W. Ted Allisona,b,c

aCenter for Prion & Protein Folding Diseases, University of Alberta,Edmonton AB, Canada; bDepartment of Medical Genetics, Universityof Alberta, Edmonton AB, Canada; cDepartment of Biological Sciences,University of Alberta, Edmonton AB, Canada

CONTACT Hadeel Alyenbaawi [email protected]

ABSTRACT

Background/Introduction: Traumatic brain injury(TBI) is known as one of the risk factors leading toneurodegeneration and dementia, in particular chronictraumatic encephalopathy (CTE). CTE is one of thetauopathies characterized by a distinct accumulationof hyperphosphorylated tau. Similar to AlzheimerDisease (AD) and other tauopathies, pathology seemsto progress to other regions during later stages of thedisease, mimicking the spreading mechanisms of priondisease. The prion-like seeding and transmission of tauin CTE and TBI cases has begun to be illuminated by alimited number of in vitro and in vivo studies.However, there is still a lack of information regardingthe mechanisms of prion-like spreading and cell uptakeof tau seeds when it comes to TBI. We aimed to estab-lish a novel TBI model using our novel transparent taubiosensor zebrafish that we engineered to detect andvisualize tau seeds. There is much promise in establish-ing in vivo models to gain greater understanding of themechanisms controlling the spreading of tau pathologyin TBI patients.Material and Methods: We successfully engineered astable transgenic zebrafish line that expresses a tauo-pathy biosensor reporter protein that detects variousforms of tau from two to four days post-transduction.We also optimized a simplified system to induce blastinjury on the tauopathy biosensor zebrafish. Zebrafishsubjected to blast injury were analysed for the forma-tion of GFP+ puncta indicative of tau seeds in thebrains and spinal cord at various time points.Additionally, we evaluated the impact of administeringconvulsant drugs as stress agents on the spreading andcell uptake of tau inclusions in both a dose- and time-dependent manner.Results: The zebrafish subjected to blast injury exhib-ited seizure-like movement immediately following theinjury. We could reliably detect GFP+ tau inclusionsthree to four days after subjecting the larvae to blastinjury. The number of these tau inclusions was signifi-cantly higher in the blast injury group compared to thecontrol group. Treatment with various concentrations

20 ABSTRACT

of convulsant drugs affected the formation of thoseinclusions in a dose-dependent manner.Conclusions: The engineering of an in vivo tau biosen-sor and TBI model in larvae zebrafish will uncoverinformation surrounding prion-like mechanisms oftau pathology and will also serve as a valuable modelfor drug screening and intervention.

45. Two prion protein paralogs of zebrafish haveopposing impacts on seizure susceptibility

Laszlo F. Locskai, Richard Kanyo, Patricia L.A. Leightonand W. Ted Allison

Centre for Prions & Protein Folding Disease, University of Alberta,Edmonton AB, Canada; Department of Biological Sciences,University of Alberta, Edmonton AB, Canada

ABSTRACT

Background: Cellular prion protein (PrPC) is famousfrom its misfolded counterpart, PrPSc, which isinvolved in neurodegenerative diseases such asCreutzfeldt Jakob’s disease. PrPC has also been linkedto Alzheimer’s disease, the most prevalent form ofdementia, by direct interactions with amyloid-β oligo-mers and amyloid-β precursor protein (APP). Ourfocus here is on understanding PrP and APP physiolo-gical interactions and functions. Reduced protein func-tions likely impact disease progression, hampering theengineering of potential therapeutics. Using the mutantzebrafish we have engineered, we seek to explore themechanism of neural regulation attributable to zebra-fish homologs of PrPC.Materials: We have engineered mutant zebrafish withdisruptions in homologs of PRNP (encoding PrPC) andAPP, being prp1−/-, prp2−/- or compound prp1−/-; prp2−/-,and appa−/-. Zebrafish larvae were treated with convul-sants pentylenetetrazol (PTZ) or kainic acid (KA) viabath treatment to induce seizures. Seizure responseswere measured by EthoVision behavioural tracking soft-ware. The neural activity of free-swimming larvae wasalso quantified during induced seizures by geneticallyencoded Calcium Modulated PhotoactivatableRatiometric Integrator (CaMPARI).Results: When induced with the convulsant PTZ, prp2−/-

mutants have a large increase in seizure response com-pared to wildtype whereas prp1−/- exhibit a more modestincrease. Neural activity in the hindbrain measured byCaMPARI presented parallel findings insomuch thatPTZ induced increased activity in prp2−/- zebrafish com-pared to wildtype. Initial results with KA suggest zebra-fish exhibit a decrease in locomotor activity in prp1mutants compared to wildtype. This contrasts com-pound mutant disruption, which appears to be similar

to locomotor activity versus wildtype. PTZ induction ofappa−/- larvae caused an increased seizure response of asimilar PTZ dose-response profile, but higher in magni-tude to prp1−/-.Conclusions: These results suggest that PrPC homologshave taken on non-additive or perhaps opposing func-tions in zebrafish neurophysiology, and that appa func-tions in neuroprotection. It appears each paralog mayregulate ion channels in opposing ways to mediate theirneuroprotective function, though further research iswarranted. Understanding which specific channels areregulated by each protein paralog, and in what fashion,has potential to illuminate the mechanism of PrPC andAPP function at the synapse.

46. Characterization of Syrian hamster adapted H-BSE prion

Kohtaro Miyazawaa, Kentaro Masujinb, YuichiMatsuuraa, Yoshifumi Iwamarua

aPrion disease unit, National Institute of Animal Health (NIAH),National Agriculture and Food Research Organization (NARO),Tsukuba, Ibaraki, Japan; bExotic Disease Research Unit, Division ofTransboundary Animal Diseases, NIAH, NARO, Kodaira, Tokyo,Japan

CONTACT Kohtaro Miyazawa [email protected]

ABSTRACT

Introduction: Bovine spongiform encephalopathy (BSE)is a prion disease in cattle, which is characterized by spon-giform changes and accumulation of a disease-associatedisoform of prion protein (PrPd). Nowadays, BSE is classi-fied into three types (classical type: C-BSE, high-type: H-BSE, and low-type: L-BSE) according to the electrophoreticmigration of their proteinase K resistant core of PrPd

(PrPres). Although Syrian hamster models have alreadyproved to be useful for studying prion diseases, transmis-sion of C- andH-BSE prions from cattle to Syrian hamstershas been inefficient. We previously reported that mouse-passaged C-BSE prion was transmissible to Syrian ham-sters. Recently, we also achieved successful transmission ofH-BSE prion from cattle to transgenic mice overexpressinghamster PrP (TgHaNSE). The distribution of immunola-beled PrPd in the brain was different between TgHaNSEmice infected with cattle H-BSE prion and those infectedwith mouse-passaged C-BSE prion even after the thirdpassage in TgHaNSE mice. However, the molecular massof the unglycosylated PrPres and glycoform profiles wereindistinguishable between them. Because these results wereobtained under artificial condition in TgHaNSE mice, weperformed a subsequent transmission of TgHaNSEmouse-passaged H-BSE prion to Syrian hamsters and comparedthe biological and biochemical properties of Syrian

PRION 21

hamster-adapted H-BSE prion to those of Syrian hamster-adapted C-BSE and L-BSE prions.Results: Syrian hamsters inoculated with 10% brain homo-genates of H-BSE prion-infected TgHaNSE mouse devel-oped the neurological signs of the disease after longincubation periods of approximately 450 days. Even afterthe second passage, hamsters survived for ~390 days. ThePrPres banding patterns and the glycoform profilesobserved in H-BSE prion-infected TgHaNSE mice weremaintained after the transmission into Syrian hamsters.Moreover, PrPres banding patterns of hamsters infectedwith H-BSE prion were identical with those of hamstersinfected with C-BSE prion even after the second passage.On the other hand, the molecular mass of the unglycosy-lated PrPres of hamsters infected with L-BSE prion wasslightly lower than those of hamsters infected with H- andC-BSE prions. Glycoform profiles were quite similarbetween hamsters infected with three different BSE prions.But the distribution of immunolabeled PrPd in the brainand the lesion profiles were different between them.Conclusion: Our data suggest that the PrPres bandingpatters and the glycoform profiles may be influencedby the properties of host PrPC rather than by prionstrain. On the other hand, biological and pathologicalfeatures including survival periods, PrPd distributionin the brain and the lesion profiles may be determinedby prion strain itself. It is interesting that the ratio ofdi-glycosylated PrPres is dominant in Syrian hamstersinfected with all three BSE prions. These modelsmight be useful for investigations of the mechanismsunderlying the conversion from PrPC to PrPSc.

47. Sugar analogues and their mechanism ofaction as prion disease therapy

Garrett J.B. Molnera, Manjeet Kumara,b and Valerie L.Sima,b

aCentre for Prions and Protein Folding Diseases, University ofAlberta; bDepartment of Medicine, Division of Neurology,University of Alberta

CONTACT Garrett J.B. Molner [email protected]

ABSTRACT

A central hallmark of prion disease is the conver-sion of normal cellular prion protein (PrPC) to apathogenic, protease resistant form (PrPSc) thatultimately leads to neurodegeneration. Speciesdevoid of prion protein are incapable of propagatingprions and therefore are incapable of prion infec-tion.1 Interestingly, pathogenic prions are hypothe-sized to exist in strains which may induce distinctstrain-specific pathologies and phenotypes. One

phenotypic strain characteristic is the relativeamounts of un, mono, and di-glycosylated PrP.2While strain information is largely thought to beencoded in the protein core conformation, there isevidence to suggest a role for glycosylation in PrPconversion and trafficking.3–5 From this concept,both a potential therapeutic strategy and an oppor-tunity to examine a role for glycosylation in strain-restricted conversion is possible.

We manipulated PrPC glycosylation in cell cultureusing naturally occurring sugar analogues and theiralky-derivatives that target α- and β-glucosidases.Treatment led to the elimination of PrPSc in CAD5 cellschronically infected with RML strain prions. However,there were concomittent changes in PrPC levels, leadingus to question whether the mechanism of action wasindeed by alteration of conversion susceptibility of PrPC(with modified sugars), or whether this treatment led to asequestration or degradation of PrPC, reducing its like-lihood of conversion. To fully understand the mechanismof inhibition, we are using immunoblot, glycoform profil-ing, and confocal microscopy to determine the effect thesecompounds have on PrP glycosylation and cellular loca-lization. Results will be discussed in the context of usingsugar analogues as a prion therapeutic.

References

[1] Büeler H, et al. Cell 1993;73(7):1339–47.[2] Khalili-Shirazi A, et al. J Gen Virol. 2005;86

(9):2635–44.[3] Makarava N, et al. Acta Neuropathol Com. 2018;6

(1):92. 4.[4] Li J, et al. Science. 2010;327(5967):869–72.[5] Katorcha E, et al. Sci Rep. 2015;5:16,912.

48. Transcriptomic analysis reveals conservedcross species functions of the cellular prion protein

Niall M Pollocka,b, Patricia L. A Leightona,b, Gavin J.Neila and W. Ted Allisona,b

aDepartment of Biological Sciences, University of Alberta,Edmonton AB, Canada; bCentre for Prion and Protein FoldingDiseases, University of Alberta, Edmonton AB, Canada

CONTACT Niall M Pollock [email protected]

ABSTRACT

Introduction: Cellular Prion Protein (PrPC) is well-stu-died due to being able tomisfold into Scrapie Prion Protein(PrPSC) causing progressive and fatal neurodegenerativediseases both in humans and other animals. Normallyfolded PrPC is implicated in the pathology of Alzheimer’sdisease, the leading cause of dementia worldwide, throughhigh affinity binding with soluble amyloid-β oligomers.

22 ABSTRACT

However, the normal function(s) of PrPC are not wellunderstood. We have used our prp1; prp2 compoundhomozygous knockout mutant zebrafish to identify poten-tially conserved, cross-species functions of PrPC usingRNA-sequencing analysis in addition to exploring theincreasing evidence for PrPC involvement in cell adhesion.Methods: Three-day post fertilization wild-type andprp1ua5003/ua5003; prp2ua5001/ua5001 zebrafish larvaeunderwent PE100-125 HiDeq2500 RNA-sequencing.Three biological replicates each consisting of 50 pooledlarvae were used for each genotype. Analysis of sequen-cing results was processed using Geneious R9/10 and Rpackages cufflinks and cummeRbund. Alterations intranscript abundance were confirmed using RT-qPCRfor selected genes of interest. Further cell adhesionstudies were carried out through in situ hybridizationanalysis and immunohistochemistry.Results: RNA-sequencing analysis of mutant fish showlarge changes in transcript abundance (log2 fold changeof 0.5 or greater) of 1,249 genes compared to wild-type fish.Of interest are genes involved in cell adhesion, includingst8sia2, ncam1a and members of the protocadherin family,which are thought to be involved during early developmentand continued maintenance of the central nervous system(CNS). Through search and comparison with publishedtranscriptomes from embryonic PrPC knockout mice, wehave identifiedmany common affected biological processesin our mutant zebrafish including apoptosis, proteolysis,oxidative stress and cell proliferation, and adhesion.Further analysis on cell adhesion through RT-qPCR ofncam1a and st8sia2 shows a significant reduction in tran-script abundance between wild-type and mutant fish, within situ hybridization of ncam1a confirming a decreaseddistribution of mRNA across the CNS.Conclusions: Our transcriptomic sequencing analysissupports a role for PrPC during early development andsustainment of the CNS, seen through significant changesin transcript abundance in members of the protocadherinfamily and other genes responsible for cell adhesion andproliferation. We propose cell adhesion is an evolutionaryconserved function of PrPC which will be of importantconsideration regarding how PrPC function may beaffected during disease.

KEYWORDS Cellular prion protein; cell adhesion;RNA-sequencing; zebrafish

49. The agent of transmissible mink encephalopathypassaged in sheep is similar to BSE-L

E. D. Cassmanna,b, S. J. Moorea,b, R. D. Kokemullera, A.Balkema-Buschmannc, M. H. Groschupc and J. J.Greenleea

aVirus and Prion Research Unit, National Animal Disease Center,ARS, United States Department of Agriculture, Ames, IA, USA (EDC,SJM, RDK, JJG); bOak Ridge Institute for Science and Education(ORISE) through an interagency agreement between the U.S.Department of Energy (DOE) and the U.S. Department ofAgriculture (USDA). ORISE is managed by ORAU under DOE con-tract number DE-SC0014664. (EDC, SJM), Department of VeterinaryPathology, Iowa State University, Ames, IA, USA (JDS); cInstitute ofNovel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut,Federal Research Institute for Animal Health, Greifswald – Isle ofRiems, Germany (ABB, MHG)

CONTACT E. D. Cassmann [email protected]

ABSTRACT

Introduction: Transmissible mink encephalopathy(TME) is a fatal neurologic prion disease of farmedmink. Epidemiologic and experimental evidence follow-ing a Wisconsin outbreak in 1985 has linked TME tolow-type bovine spongiform encephalopathy (BSE-L).Evidence suggests that farmed mink were likely exposedthrough feeding of BSE-L infected downer cattle. Theinterspecies transmission of TME to cattle has beendocumented. Recently, we demonstrated the susceptibil-ity of sheep to cattle passaged TME by intracranialinoculation. The aim of the present study was to com-pare ovine passaged cattle TME to other prion diseasesof food-producing animals. Using a bovine transgenicmouse model, we compared the disease phenotype ofsheep TME to BSE-C and BSE-L.Materials and Methods: Separate inoculants of sheeppassaged TME were derived from animals with theVRQ/VRQ (VV136) and ARQ/VRQ (AV136) prion pro-tein genotype. Transgenic bovinized mice (TgBovXV)were intracranially inoculated with 20 µl of 1% w/vbrain homogenate. The disease phenotypes were char-acterized by comparing the attack rates, incubationperiods, and vacuolation profiles in TgBovXV mice.Results: The attack rate for BSE-C (13/13), BSE-L (18/18), and TMEVV (21/21) was 100%; whereas, theTMEAV group (15/19) had an incomplete attack rate.The average incubation periods were 299, 280, 310, and541 days, respectively. The vacuolation profiles of BSE-L and TMEVV were most similar with mild differencesobserved in the thalamus and medulla. Vacuolationprofiles from the BSE-C and TMEAV experimentalgroups were different than TMEVV and BSE-L.Conclusion: Overall the phenotype of disease in TMEinoculated transgenic mice was dependent on thesheep donor genotype (VV vs AV). The results ofthe present study indicate that TME isolated fromVRQ/VRQ sheep is similar to BSE-L with regardsto incubation period, attack rate, and vacuolationprofile. Our findings are in agreement with previousresearch that found phenotypic similarities between

PRION 23

BSE-L and cattle passaged TME in an ovine trans-genic rodent model. In this study, the similaritiesbetween ovine TME and BSE-L are maintained aftermultiple interspecies passages.

50. Cellular Prion protein on human leucocytes isassociated with iron metabolism

Ewald Lindnera, Nora Woltschea, DomagojIvastinovica, Verena Zrimb, Rita Moosc, AndreasWedricha and Adriano Aguzzic

aDepartment of Ophthalmology, Medical University Graz; bCoreFacility Imaging, Center for Medical Research Graz; cInstitute ofNeuropathology, University Hospital Zurich

CONTACT Ewald Lindner [email protected]

ABSTRACT

Introduction: The function of the prion proteinremains elusive. Several findings indicate a role iniron metabolism. In this study, we wanted to investigatean association between indicators of iron storage andthe expression of prion protein on human leucocytes inglaucoma patients.Materials and Methods: Patients were recruited atthe Department of Ophthalmology at the MedicalUniversity Graz. Twenty patients with glaucomawere included. The expression of prion protein onCD3+ CD4+, CD3+ CD8+ and CD14+ leukocyteswas investigated by FACS with POM1 antibody.Results: The expression of Prion Protein correlatedsignificantly with soluble transferrin receptor, haemo-globin, and serum iron (Pearson r > 0.6; p-value <0.01). No association was found between expression ofprion protein and glaucoma progression.Conclusions: We found a strong correlation betweeniron metabolism and the expression of prion protein onhuman leucocytes. This presents add to our under-standing of the function and regulation of the cellularprion protein.

KEYWORDS: Prion protein; human leucocytes; solu-ble transferrin receptor; haemoglobin

51. Cryptic prion strains/variants appear whenanti-prion systems are disabled

Reed B. Wickner, Moonil Son, Evgeny Bezsonov,Songsong Wu, Morgan Dewilde and Herman K. Edskes

Laboratory of Biochemistry and Genetics, National Institute ofDiabetes and Digestive and Kidney Diseases, National Institutes ofHealth, Bethesda, MD, USA

CONTACT Reed B. Wickner [email protected]

ABSTRACT

The yeast prions [PSI+] and [URE3] are in-registerparallel amyloids of Sup35p and Ure2p, respectively.Ssb1,2p are ribosome-associated Hsp70s that ~10-foldrepress [PSI+] generation. Upf1,2,3p are factorsessential for mRNA nonsense-mediated decay that,at normal levels, cure >90% of [PSI+] variants arisingin any upf mutant. The formation of the Upf1,2,3-Sup35 complex is critical for the curing process. Theupf mutants show 10–15 fold elevation in frequencyof spontaneous or induced [PSI+] generation. Bycontrolling cellular levels of 5-pyrophospho-inositolpentaphosphate, Siw14p prevents the propagation ofabout half of the [PSI+] variants arising in itsabsence. Normal levels of the disaggregating chaper-one Hsp104 cures about half of the [PSI+] variantsthat arise in an hsp104T160M mutant defective in theHsp104 overproduction curing activity. Normal levelsof Btn2p and Cur1p are paralogs that cure ~90% of[URE3] prion strains/variants arising in a btn2Δcur1Δ strain, preferentially those with low propagonnumbers. Btn2p (but not Cur1p) cures by collectingUre2p amyloid filaments at a single site in the cellco-inciding with Btn2p itself. Cell division results infrequent curing of one of the daughter cells.

The common thread of most of these systems is thatprion strains that would not survive in a wild typestrain are detected in mutants defective in one ormore of these anti-prion systems. We infer that thenormal cell experiences a large load of nascent prions,most of which are quickly eliminated by one or more ofthese systems. The mechanistic details, insofar as weknow them, argue that each of these systems are dis-tinct. The excess prion strains/variants detected in cellsdefective in one anti-prion system are evidentlyimmune to curing by the other anti-prion systems,indicating that each anti-prion system defines a newclass of prion strains/variants. It is likely that someprion/prion-like diseases are a consequence of break-down of homologous/analogous human anti-prionsystems.

Future work will address the following questions:

● What other components are involved in each ofthese processes? Is there any overlap?

● Do cells defective in multiple anti-prion systemsshow even higher prion generation frequencies?

● What other anti-prion systems are there?

24 ABSTRACT

52. Microglial-specific exosome isolation fromprion-infected fluids

Stephanie Booth, Lise Lamoureux and Sarah Medina

Public Health Agency of Canada, NML

ABSTRACT

Prion disease is caused by the conversion of PrPc (cel-lular prion protein) to PrPsc (abnormal prion protein)which aggregates in the brain causing spongiform neu-rodegeneration. In response, astrocytes and microgliaare activated. Both PrPc and PrPsc have been shown tobe associated with exosomes, which are thought to beinvolved in prion pathogenesis. Exosomes are small 30–150nm vesicles that are released from cells via exocy-tosis into biological fluids. They are involved in cell tocell communication as they can cross biological barrierseasily. Exosomes contain protein, lipids, and nucleicacids that are representative of the cell types fromwhich they originate. Therefore, they could be specifi-cally isolated and used as the basis for cell-type specificbiomarker discovery. Few studies have looked at prion-specific changes in the expression of biomolecules inexosomes from cells that are involved in the diseaseprocess. Exosomes originating from the brain (specifi-cally microglia) could be an ideal source for disease-specific molecular signatures from early stages of dis-ease. We hypothesized that TMEM119, a transmem-brane protein that is expressed highly specific onmicroglial cells, could be used as a marker to specifi-cally select microglial exosomes. The goal of this pre-liminary study was to develop methods to isolatemicroglial-specific exosomes from cells infected withprions. We used western blots and nanoparticle track-ing analysis (NTA) to look at the composition ofmicroglial-specific exosomes. Exosomes were isolatedin media from BV2 cells (a microglial cell line). ATMEM119 antibody was labelled with a fluorophoreand binding to exosomes was visualized via NTA.Exosomes were also isolated from prion-infected anduninfected primary cerebellum slice cultures.TMEM119 antibody was bound to beads which couldthen successfully ‘pull-out’ microglial specific exo-somes. This methodology will be tested for its utilityin prion disease-specific biomarker discovery frommicroglial exosomes isolated from biofluids frominfected animals.

53. Evaluation of the inter-species transmissionpotential of different CWD isolates

Rodrigo Moralesa, Carlos Kramma,b, Paulina Sotoa,Adam Lyona, Sandra Pritzkowa, Claudio Sotoa

aMitchell Center for Alzheimer’s disease and Related BrainDisorders, Dept. of Neurology, McGovern School of MedicineUniversity of Texas Health Science Center at Houston, TX, USA;bFacultad de Medicina, Universidad de los Andes, Santiago, Chile

ABSTRACT

Chronic Wasting Disease (CWD) has reached epidemicproportions in North America and has been identified inSouth Korea and Northern Europe. CWD-susceptiblecervid species are known to share habitats with humansand other animals entering the human food chain. Atpresent, the potential of CWD to infect humans andother animal species is not completely clear. Theexploration of this issue acquires further complexityconsidering the differences in the prion proteinsequence due to species-specific variations and poly-morphic changes within species. While several speciesof cervids are naturally affected by CWD, white-taileddeer (WTD) is perhaps the most relevant due to itsextensive use in hunting and as a source of food.Evaluation of inter-species prion infections using ani-mals or mouse models is costly and time consuming. Weand others have shown that the Protein MisfoldingCyclic Amplification (PMCA) technology reproduces,in an accelerated and inexpensive manner, the inter-species transmission of prions while preserving thestrain features of the input PrPSc. In this work, we testedthe potential of different WTD-derived CWD isolates totransmit to humans and other animal species relevantfor human consumption using PMCA. For these experi-ments, CWD isolates homozygous for the most commonWTD-PrP polymorphic changes (G96S) were used (96SSvariant obtained from a pre-symptomatic prion infectedWTD). Briefly, 96GG and 96SS CWD prions wereadapted in homologous or heterologous substrate byPMCA through several (15) rounds. End products, aswell as intermediates across the process, were tested fortheir inter-species transmission potentials. A similarprocess was followed to assess seed-templated misfold-ing of ovine, porcine, and bovine PrPC. Our results showdifferences on the inter-species transmission potentialsof the four adapted materials generated (PrPC/PrPSc

polymorphic combinations), being the homologouscombinations of seed/substrate the ones with the greaterapparent zoonotic potential. Surprisingly, 96SS prionsadapted in homologous substrate were the ones showingthe easiest potential to template PrPC misfolding fromother animal species. In summary, our results show thata plethora of different CWD isolates, each comprisingdifferent potentials for inter-species transmission, mayexist in the environment. These experiments may help toclarify an uncertain and potentially worrisome publichealth issue. Additional research in this area may be

PRION 25

useful to advise on the design of regulations intended tostop the spread of CWD and predict unwanted zoonoticevents.

54. Analysis of amyloid properties of MBP proteinin the brain of rat R. norvegicus

A. A. Shenfelda,b*, M. J. Velizhaninaa, J. V. Sopovaa,b, T.A. Belashovab and A. P. Galkina,b

aSt-Petersburg State University, dept of Genetics andBiotechnology, St-Petersburg, Russia; bSt-Petersburg branch ofVavilov Institute of General Genetics, Lab. of Genetic Modeling ofHuman Diseases, St-Petersburg, Russia

CONTACT A. A. Shenfeld [email protected]

55. PAD-beads enhances detection of PrPSc inscrapie infected sheep brain samples using real-time quaking-induced conversion

Soyoun Hwang, Rohana P. Dassanayake and Eric M.Nicholson

United States Department of Agriculture, Agricultural ResearchService, National Animal Disease Center, Ames, Iowa, USA

CONTACT Soyoun Hwang [email protected]

ABSTRACT

Scrapie, a transmissible spongiform encephalopathy(TSE), naturally occurs in sheep and goats. This fatalneurodegenerative disease results from misfolding ofthe cellular prion protein (PrPC) to a pathogenicprion protein form (PrPSc). This pathogenic form accu-mulates in the brain and other tissues.The presence ofPrPSc can been detected by an in vitro conversion assayknown as real-time quaking induced conversion (RT-QuIC). RT-QuIC has been used to detect PrPSc in avariety of biological tissues from brains to fluids. Whilethis technique is both rapid and sensitive enhancing thedetection of prions would be valuable in the diagnosticlaboratories. In this study, we evaluated if RT-QuIC canbe coupled to PrPSc binding to a PrPSc specific ligandfound on commercially available PAD-Beads for cleanup of PrPSc containing samples enhanced prion detec-tion. Coupling the RT-QuIC amplification to PAD-Bead based cleanup allowed detection of scrapie prionrapidly and without dilutions sheep brain homogenatesprior to RT-QuIC. The PAD-Bead sample pretreatmentstep prior to RT-QuIC is a useful enhancement in thediagnosis of TSEs.

KEYWORDS: Scrapie; prion diseases; RT-QuIC; trans-missible spongiform encephalopathy; PAD-Beads; mag-netic particle extraction

56. Understanding chronic wasting disease spreadpotential for at-risk species

Catherine I. Cullingham, Anh Dao, Debbie McKenzieand David W. Coltman

Department of Biological Sciences, University of Alberta, EdmontonAB, Canada

CONTACT Catherine I. Cullingham [email protected]

ABSTRACT

Genetic variation can be linked to susceptibility or resis-tance to a disease, and this information can help to betterunderstand spread-risk in a population. Wildlife diseaseincidence is increasing, and this is resulting in negativeimpacts on the economy, biodiversity, and in someinstances, human health. If we can find genetic variationthat helps to informwhich individuals are susceptible, thenwe can use this information on at-risk populations to bettermanage negative consequences. Chronic wasting disease, afatal, transmissible spongiform encephalopathy of cervids(bothwild and captive), continues to spread geographically,which has resulted in an increasing host-range. The diseaseagent (PrPCWD) is a misfolded conformer of native cellularprotein (PrPC). In Canada, the disease is endemic inAlberta and Saskatchewan, infecting primarily mule deerandwhite-tail deer, with a smaller impact on elk andmoosepopulations. As the extent of the endemic area continues toexpand, additional species will be exposed to this disease,including bison, bighorn sheep,mountain goat, and prong-horn antelope. To better understand the potential spread-risk among these species, we reviewed the current literatureon species that have been orally exposed to CWD to iden-tify susceptible and resistant species. We then comparedthe amino acid polymorphisms of PrPC among these spe-cies to determine whether any sites were linked to suscept-ibility or resistance to CWD infection. We sequenced theentire PrP coding region in 578 individuals across at-riskpopulations to evaluate their potential susceptibility. Threeamino acid sites (97, 170, and 174; human numbering)were significantly associated with susceptibility, but thesewere not fully discriminating. All but one species amongthe resistant group shared the same haplotype, and thesame for the susceptible species. For the at-risk species,bison had the resistant haplotype, while bighorn sheepand mountain goats were closely associated with the resis-tant type. Pronghorn antelope and a newly identified hap-lotype in moose differed from the susceptible haplotype,but were still closely associated with it. These data suggestpronghorn antelope will be susceptible to CWD whilebison are likely to be resistant. Based on this data, recom-mendations can bemade regarding species to bemonitoredfor possible CWD infection.

26 ABSTRACT

KEYWORDS: Chronic wasting disease; Prnp; wildlifedisease; population genetics; ungulates

57. Oligomerization of Aβ(1–40/42) in the presenceof somatostatin-14 investigated in silico

Min Wua, Lyudmyla Dorosha,b, Gerold Schmitt-Ulmsc,Holger Willed,e, Maria Stepanovaa,b

aDepartment of Electrical and Computer Engineering, University ofAlberta, Edmonton, Canada; bNational Research Council of Canada,Edmonton, Alberta, Canada; cLaboratory Medicine andPathobiology, University of Toronto, Toronto, Canada;dDepartment of Biochemistry, University of Alberta, Edmonton,Canada; dCentre for Prions and Protein Folding Diseases,University of Alberta, Edmonton, Canada

CONTACT Min Wu [email protected]; Lyudmyla Dorosh [email protected]

ABSTRACT

Alzheimer’s disease (AD) is associated with pathologicaloligomeric aggregates and fibrils formed by amyloid beta(Aβ) peptides. Experiments indicated that somatostatin-14(SST14), a small cyclic peptide found in the brain, mayinfluence the formation of toxic oligomers of Aβ1–42, butnot Aβ1-40 [1]. However, the specific role of SST14 and thereason for the different responses of Aβ42 and Aβ40 remainunclear. We have investigated the effects of SST14 onaggregation of Aβ1-40 and Aβ1–42 peptides using 100 nslong all-atom molecular dynamics simulations. Startingfrom randomly positioned Aβ40/42 and SST14 chains inwater, we observed the formation of oligomers, and inves-tigated their structure and dynamics. For comparison, wehave investigated a similar system containing Aβ1–42 andthe arginine vasopressin (AVP) peptide as a negative con-trol [1]. We have employed well-established structuralbiology tools, as well as a novel essential collectivedynamics (ECD) method [2], which allows analysing paircorrelations, main-chain flexibilities, and domains of cor-related motion within the same framework. We observedformation of stable co-oligomers containing Aβ chains andSST14 or AVP molecules in all the three systems, Aβ42-SST14, Aβ40-SST14, andAβ42-AVP. During the simulations,the number of hydrogen bonds in the systems graduallyincreased. This was accompanied by a buildup of saltbridges, hydrophobic contacts, and other interactionsresponsible for the oligomerization process. Interestingly,C-terminal residues of both Aβ42 and Aβ40 are oftenlocated at the surface of the oligomers, whereas their cen-tral parts stay preferentially in the interior regions. TheECD analysis indicated that in most Aβ oligomers, C-termini exhibited greater main-chain flexibility in compar-ison to central regions, also indicating a stronger solvent

exposure of the C-termini. The solvent accessible surfacearea (SASA) of hydrophobic side groups is the greatest inAβ42-SST14 oligomers, in part due to the larger hydropho-bic SASA of SST14 in comparison to the AVP control.Aβ42-AVP oligomers are relatively small in size, and theirhydrophobic SASA is less than in the other systems. Weconclude that differences in aggregation of Aβ42-SST14 andthe other two systems may result from a greater hydro-phobic SASA of both Aβ42 and SST14, producing a stronger‘sticky surface’ effect. In Aβ42-AVP the effect appears les-sened by a lower hydrophobicity of AVP, whereas in Aβ40-SST14, the reduced number of solvent-exposed hydropho-bic C-terminal residues of Aβ may play a role.

References

[1] Wang H, Muiznieks LD, Ghosh P, et al. Somatostatinbinds to the human amyloid β peptide and favors theformation of distinct oligomers. eLife. 2017;6:e28401.

[2] Dorosh L, Stepanova M. Probing oligomerization ofamyloid beta peptide in silico. Mol.BioSyst.2017;13:165–182.

58. Understanding the mechanisms and effects ofimpaired Rab7 function in prion infection

Pearl Cherry, Su Yeon Shim and Sabine Gilch

Department of Ecosystem and Public Health, Calgary PrionResearch Unit, Faculty of Veterinary Medicine; Hotchkiss BrainInstitute; University of Calgary, Calgary, Canada

CONTACT Pearl Cherry [email protected]

ABSTRACT

Background: In persistently prion-infected cell cultures,reduced membrane association of Rab7 and, as a conse-quence, reduced lysosomal maturation and impaired lyso-somal degradation is observed. Other hallmarks of prioninfections are increased cholesterol levels and impairedaxonal retrograde trafficking. Targeted cargo trafficking isregulated mainly by rab proteins and requires them toshuttle between an active GTP and membrane bound andan inactive cytosolic state. Rab7 is a protein essential forintracellular transport of cargos like low density lipoprotein(LDL) and neurotrophins. The aim of this study is toelucidate the causes and effects of reduced membranebound Rab7, specifically on how it influences PrPSc propa-gation, trafficking of low density lipoprotein (LDL) andneurotrophins and hence its effect on cholesterol metabo-lism and neurodegeneration.Methods: We used prion infected (strain 22L) N2a cells fortransient transfection with different functional mutants ofRab7 including thewild type (WT), the constitutively active

PRION 27

(Q67L) and the transdominant negative (T22N) mutantsfor 48 h and analysed PrPSc levels. The differential effect ofoverexpression of thesemutants on cholesterol metabolismin 22LN2a cells is analysed through Amplex cholesterolassay. Immunofluorescence experiments are conductedusing an antibody against Rab7-GTP in prion infectedand non-infected cell lines. Trafficking of LDL is analysedusing by pulse-chase experiments with fluorescentlylabelled LDL and co-localization with endosomal markers.

Results: We demonstrate that in the prion infected celllines, transient over-expression of the constitutively activemutant Rab7 reduces PrPSc propagation. Since prioninfected cell cultures exhibit an increase in the cholesterollevels, we analysed how different functional mutants ofRab7 regulate cholesterol levels and observed that over-expression of the constitutively active Q67L mutantreduces the cholesterol level with respect to Rab7-WTmutant in 22LN2a cells. Overexpression of Rab7-WT didneither affect PrPSc levels, nor cholesterol content in thosecells. This indicates that activation of Rab7 by GDP/GTPexchange might be impaired in 22LN2a cells. Preliminaryresults of a comparative analysis of Rab7-GTP in N2a and22LN2a point at a reduction of Rab7-GTP upon prioninfection. Since Rab7 is required for LDL transport, LDLtrafficking is analysed to define a link between aberrantvesicle trafficking and enhanced cholesterol synthesis.Conclusion: Through this study, we shed light on theunderlyingmechanisms and effects of impairedmembraneassociation of Rab7 in prion infection. These effects includedeterioration of cellular processes such as vesicle traffickingand cholesterol metabolism, hence favouring PrPScpropagation.

59. The role of glial cells in BSE pathology: ahistomorphological and immunohistochemicalinvestigation

Christine Fasta, Kristina Santiago-Mateob, RakhiKatochb, Martin H. Groschupa, Ute Zieglera, StefanieCzubb

aFriedrich-Loeffler Institut, Isle of Riems, Germany; bCanadian FoodInspection Agency, Lethbridge, Alberta, Canada

CONTACT Christine Fast [email protected]

ABSTRACT

Background/Introduction: Bovine spongiform encepha-lopathy (BSE) is a fatal neurological disease of cattle asso-ciated with the accumulation of an altered form of thecellular prion protein, termed pathological prion protein(PrPBSE) The pathomechanisms linking abnormal proteinaccumulation with the typical severe neurological altera-tions in BSE are less than clear. Here we investigated the

role of glia cells in the pathology of BSE infected cattle,specifically if and to which degree the histopathologicalchanges are glia mediated and therefore due to the hostresponse and not to the BSE agent itself.Material and Methods: We examined 29 experimen-tally BSE infected cattle at preclinical (no PrPBSE in thebrain stem), late preclinical (no clinical signs, firsttraces of PrPBSE in the brain stem), and clinical (clinicalsigns and distinct accumulation of PrPBSE in the brainstem) stages of the disease. The following definedregions of the CNS were examined: Obex, RedNucleus, Cerebellum, Parietal Cerebrum, SeptalNucleus, and Hippocampus and both histopathologicallesions and PrPBSE accumulation were determined.Additionally, antibodies Iba-1, CX3CR1, and GFAPwere used to visualize and identify glial involvementwithin the context of the disease. In addition, fourunchallenged animals of the same age groups wereincluded as negative controls.Results: PrPBSE is found only in neurons and microglia.Different analytic approaches showed that the mostconsistent changes in glial numbers and phenotypeswere detected in the Obex, Red Nucleus, andHippocampus. However, only in clinical affected cattle,we observed with all antibodies a mild diffuse increasein glial reactivity, indicating a gliosis, as compared tocontrols, preclinical, and late preclinical animals.Additionally, different glial reaction patterns werealmost exclusively detected in clinical animals. Thisincludes a hyperplastic and in parts irregular appear-ance of the glial network, an accumulation of clumsycells with shortened processes and rounded cell bodies,a diffusely distributed coarse granular staining andoccasionally perineuronal cluster of glial cells.

Conclusion: Apart from neurons, we identifiedmicroglia as the only other PrPBSE accumulating cellpopulation; computerized assessment of IBA1,CX3CR1, and GFAP antibodies are ongoing. Thepreliminary results presented here indicate that theactivation of a glial response is a rather late event,most probably associated with the accumulation ofPrPBSE.

60. Caribou Prnp polymorphism distribution inCanada and its impact on CWD pathogenesis

Maria I. Arifina, Samia Hannaouia, Yuan-Hung Huanga,Gordon Mitchellb, Antanas Staskeviciusb, LechKaczmarczykc,d, Walker Jacksonc,d and Sabine Gilcha

aDepartment of Ecosystem and Public Health, University of Calgary;bNational and OIE Reference Laboratory for Scrapie and CWD, OttawaLaboratory Fallowfield, Canadian Food Inspection Agency;cWallenberg Center for Molecular Medicine, Department of Clinical

28 ABSTRACT

and Experimental Medicine, Linköping University; dGerman Center forNeurodegenerative Disease (DZNE), Bonn

CONTACT Maria I. Arifin [email protected]

ABSTRACT

Wild reindeer in Norway were recently diagnosed withchronic wasting disease (CWD) [1]. Prions from CWD-infected deer were also transmissible to other reindeer inexperimental settings [2,3]. Although CWD has not yetbeen reported in wild reindeer (caribou) in Canada, thesestudies show that they are at risk of infection. Thewild-typereindeer, homozygous for serine at prion protein (PrP)residue 138 (138SS), developed clinical disease upon oralinoculation. Animals carrying at least one asparagine allele(138SN, 138NN) accumulated prions only in the periphery.However, both genotypes were susceptible to intracerebralprion inoculation [2,3]. Fallow deer are wild-type 138NNand are resistant to peripheral but not intracerebral prioninfection [4]. Thus, we hypothesize that the138N allele,present in caribou herds in Alberta [5], alters CWD patho-genesis by limiting prion transport from the periphery tothe CNS. Our aim is to elucidate the mechanisms involvedin this process.

We extracted DNA from ±800 caribou, sequenced thePrnp open reading frame and determined the 138N alleleprevalence in Canadian caribou populations. Resultsshow that the 138N allele was highly prevalent in theChinchaga woodland caribou population in BC (64%). Itwas also higher in barren-ground (37%) than in otherwoodland caribou populations (26%). To analyse howthe 138N allele affects CWD pathogenesis, we generatedknock-in (KI) mice where the mouse PrP is replaced bywild-type 138SS or 138NN cervid PrP. The KIs wereobtained by injecting CRISPR/Cas9-edited C57BL6embryonic stem cells (Bruce4) into wild-type blastocysts,generating chimeras, and breeding progeny to homozyg-osity in a C57BL6 background. PrP expression was deter-mined using western blotting and qPCR. Correct PrPpost-translational modifications were confirmed byPNGase-F and Endo-H digestion. We will inoculate ourKIs with CWD-positive material from the correspondingreindeer genotypes [1,2]. The CWD-infected reindeermaterial was characterized by real-time quaking-inducedconversion (RT-QuIC) and its transmissibility to trans-genic mice overexpressing elk PrP (TgElk). Attack rate inthe TgElks were almost 100%, except for those inoculatedwith 138SN lymph node material. Western blotting con-firmed the presence of proteinase-K resistant PrP in allterminally ill mice. Our goal is to analyse the susceptibilityof KIs carrying the 138N allele to intracerebral and per-ipheral prion infection. We will also assess the efficiencyof prion transport from the periphery to the CNS in

intraperitoneally inoculated KIs. Prion strain propagationwithin a host is highly dependent on its PrP genotype.This study will provide insight into how the 138N PrPspecifically influences CWD propagation in caribou.

KEYWORDS: Chronic wasting disease; polymorphism;caribou; reindeer; cervid; transgenic mice; knock-inmice

References

[1] Benestad, et al. Vet Res. 2016;47:1.[2] Mitchell, et al. PLoS ONE. 2012;7(6):e39055.[3] Moore, et al. Emerg Infect Dis 2016;22(12):2142–5.[4] Rhyan, et al. J Wildl Dis. 2011;47(3):739–4.[5] Cheng, et al. Prion. 2017;11(2):136–42.

61. Prion propagation in cultured astrocytes: amodel for prion infection in astroglia

Waqas Tahira, Basant A Abdulrahmana, Dalia HAbdelaziza, Simrika Thapaa, Rupali Waliaa andHermann M Schatzla

aDepartment of Comparative Biology & Experimental Medicine,Faculty of Veterinary Medicine, University of Calgary, Calgary,Alberta, Canada; bCalgary Prion Research Unit, and HotchkissBrain Institute, University of Calgary, Calgary, Alberta, Canada

CONTACT Waqas Tahir [email protected]

ABSTRACT

Background: Neuronal cell lines permissive to prioninfection are useful experimental tools for studying themolecular mechanisms underlying prion infection.Such cell models are widely used to find and character-ize chemical compounds which have anti-prion poten-tial. Unfortunately, findings from cell culture are notvery predictive for in vivo efficacy of such compounds.One reason might be that other cell types in the brainpropagate prions, and that their processing of prionsmight differ from that of neurons. Exactly how astro-cytes propagate, clear and disseminate prions is notwell studied, explained by the lack of astrocyte cellmodels permissive of stable prion infection.Objective: We investigated the permissiveness of animmortalized and uncloned astrocyte cell line to prionpropagation, in order to develop a persistent prioninfection model of astrocytes.Material and Methods: We quantified mRNA and cellsurface levels of PrPC in astrocytes with qPCR and FACSanalysis, respectively, and compared them to those of otherpermissive neuronal (N2a and CAD5) and non-neuronal(MEF) cell lines. Next, we inoculated immortalized astro-cytes with three mouse-adapted scrapie prion strains (22L,

PRION 29

RML and ME7), and cultured them continuously for mul-tiple passages. Prion propagation was examined withimmunoblotting, RT-QuIC, and immunofluorescencemethods at every passage.Results: Surface expression of PrPC was comparablebetween astrocytes and N2a and CAD5 cells, and washigher than in MEF cells. Astrocytes infected with 22Lprions showed PrPSc in immunoblot analysis through-out all tested passages. Similarly, immunofluorescenceresults showed that astrocytes were able to propagate22L prions stably. Infection with Me7 prions was nega-tive in immunoblot, with transient weak signals inimmunofluorescence and RT-QuIC analysis. AlthoughRML infection was negative in immunoblot until pas-sage 6 p.i., RML-infected astrocytes had the strongestsignal in RT-QuIC in all passages (p1-p6).Conclusions: Our data show that this uncloned astro-cyte cell line is permissive to stable prion infection,depending on the prion strain. Whereas infection with22L prions resulted in the expected strong signals inimmunoblot, RT-QuIC, and immunofluorescence ana-lysis, infection with RML prions yielded a stable infec-tion which was positive in RT-QuIC and mostlynegative in immunoblot analysis. Obviously, RML and22L prions were handled very differently by this cellline, with regard to prion conversion and PK resistance.This new cell model will be helpful for better under-standing of the role of astrocytes in prion infection.

62. A novel ex vivo model for characterization ofAlzheimer’s disease strains

Hailey E. Pineaua,b, Grant Normana,b, DavidWestawaya,b and Valerie L. Sima,b

aCentre for Prions and Protein Folding Diseases, University ofAlberta; bDepartment of Medicine, Faculty of Medicine &Dentistry, University of Alberta

CONTACT Hailey E. Pineau [email protected]

ABSTRACT

Background: Affecting >48 million people worldwide,Alzheimer’s disease (AD) is the leading cause of dementia.Evidence suggests that both amyloid β (Aβ) and tau, theproteins associated with neurodegeneration in AD, existin many different conformations (strains), which mayexplain variation in pathology and phenotype betweenAD patients. Specifically, we are interested in whetherstrain differences underlie differences in rate of diseaseprogression. Using biophysical techniques and prion-based slice culture assays, we hope to elucidate Aβ proper-ties and plaque morphology characteristic of typical and

rapidly progressive AD, and to develop a novel ex vivomodel that can replicate strain features.Methods: The organotypic slice culture assay (OSCA) is amouse brain slice culture that can be infected with differ-ent strains of prions and undergoes pathology as seen invivo. Because Aβ and tau possess prion-like infectivity, weare adapting this technique to AD using coronal brainslice cultures from 5xFAD and CRND8 mice. These miceexpress human mutant amyloid precursor protein anddevelop plaques and other neuropathology by 2 and3 months of age, respectively. Typical or rapidly progres-sive AD brain homogenate will be applied to these slicecultures to induce strain-specific aggregates. Using con-focal microscopy, we will quantify plaque burden, mor-phology, localization, and adjacent pathology. ELISA willbe done to quantify levels of Aβ40 and Aβ42 in the ADbrain samples and slice culture. Guanidinium denatura-tion curves will allow for comparison of Aβ conforma-tional stability between typical and rapidly progressiveAD brain samples. Additionally, real-time quakinginduced conversion (RT-QuIC) will be performed toassess seeding ability and aggregation kinetics of the Aβin all of the AD samples.Results: Confocal images of Aβ plaques stained withthioflavin S and/or 6E10 in fresh and cultured sliceswill be compared, and the effects of exposure to rapidlyprogressive AD brain homogenate will be shown.ELISA results will demonstrate a positive correlationbetween disease duration and hippocampal levels ofAβ42 and RT-QuIC data on aggregation kinetics mayalso be presented.Conclusions: An ex-vivo model of AD that canreplicate strain features will facilitate investigationinto AD pathogenesis and strain-specific treatments.

63. Alteration of prion strain emergence by non-host factors

Sara A. M. Holec and Jason C. Bartz

Department of Medical Microbiology and Immunology, CreightonUniversity School of Medicine, Creighton University, Omaha,Nebraska, USA

CONTACT Sara A. M. Holec [email protected]

ABSTRACT

Prions can persist in the environment for extendedperiods of time after adsorption to surfaces includingsoils, feeding troughs, or fences. The adsorption ofPrPSc to surfaces is influenced by a several factorsincluding the prion strain, species of origin, and surface

30 ABSTRACT

type. Attachment to surfaces can alter PrPSc properties.The cumulative effect of these factors can result instrain- and soil-specific differences in prion adsorption,infectivity, and response to inactivation. Environmentalprocesses such as wetting and drying or freezing andthawing can decrease PrPSc conversion efficiency andenhance PrPSc degradation. Little is known about howstrain-specific incomplete inactivation of surface boundPrPSc affects transmission and strain emergence. Toexamine this biologically relevant mode of prion trans-mission, we investigated the effect of surface bindingand environmental forces on the emergence of a strainfrom a mixture.

To investigate the effects of environmental forces onstrain emergence, we tested multiple parameters includ-ing proteolytic digestion, surface adsorption, and repeatedcycles of wetting and drying. Extensive proteinase K (PK)digestion of HY and DY PrPSc resulted in earlier emer-gence of HY PrPSc in protein misfolding cyclic amplifica-tion strain interference (PMCAsi) compared toundigested controls. This result confirmed the proof ofprinciple that selective alteration of the starting ratios ofconversion competent HY and DY PrPSc can alter strainemergence. We then further investigated if environmen-tally relevant factors such as surface binding and weath-ering could have a similar effect. Adsorption of HY andDY PrPSc to silty clay loam (SCL) reduces conversionefficiency that can be strain-dependent; however, adsorp-tion of HY and DY PrPSc to SCL, both individually andtogether, did not alter strain emergence compared tounbound control PMCAsi reactions. Similarly, repeatedcycles of wetting and drying of HY and DY PrPSc boundto SCL did not alter the emergence of HY PrPSc. This datasuggests that prion strain interference can occur whenbound to surfaces. Since it is likely that surface boundPrPSc is responsible for a significant portion of priontransmission under natural conditions, this is a highlysignificant finding. Under the limited conditions tested,we found that surface binding and weathering did notalter the emergence of a HY PrPSc.

64. Investigation of the role of lamin A-inducedmolecular ageing in tauopathy

Zhuang (George) Han AND David Westaway

Centre for Prions and Protein Folding Diseases, Department ofBiochemistry, Faculty of Medicine and Dentistry, University ofAlberta

CONTACT Zhuang (George) Han [email protected]

ABSTRACT

Tauopathies are a diverse group of age-related neuro-degenerative diseases characterised by filamentous Tau

lesions in brain [1]. Alzheimer’s disease, frontotem-poral dementia and progressive supranuclear palsy areexamples of tauopathies that have presented a tremen-dous challenge to health care systems worldwide [2–4].Tau is a microtubule-associated protein predominantlyexpressed in axons of neurons. In disease states, itbecomes hyperphosphorylated and migrates to the cellbody, forming aggregates that are toxic to neurons[1,5]. Certain tauopathies may associate with impairedlamin-enriched nuclear lamina and thus are also con-sidered laminopathies, at least in the experimental con-text of Drosophila melanogaster [6,7].

Hutchinson Gilford progeria syndrome (HGPS), anautosomal dominant disorder of premature ageing, associ-ates with a truncated form of lamin A (termed ‘progerin’)[8,9]. Neurons in the brain mainly produce abundantlamin C but little lamin A, a consequence of prelamin AmRNA degradation by a brain-specific microRNA [10].Although this reasonably explains the absence of neurolo-gical symptoms in lamin A-induced accelerated ageing, itremains unclear whether progerin – if it were expressed inneurons – would enhance pathological Tau-induced neu-rotoxicity in brain, given that overexpression of progerincan be toxic to enteric nerves and stem cell-derived neu-rons [11,12]. To address this question, we are using theHGPS progeric mutation of lamin A to accelerate cellularageing in petri-dish cultures; we hypothesise that when theexpression level of aggregation-prone Tau is low (near-physiological level), lamin A- induced cellular changesmight amplify or alter its pathogenic potential.

Progeric and wildtype forms of lamin A were trans-fected into an HEK293 Tau RD-YFP reporter cell line[13] with subcellular localization of progerin and perturba-tion of the nuclear lamina assessed by immunohistochem-istry. Following transduction of different classes of Taufibril derived from transgenic P301L mice and humanP301L patients, morphologies of Tau aggregates in HEKreporter cells with and without lamin A expression aredetermined by fluorescence microscopy. Aggregated Tauis then harvested, digested with trypsin and analysed withcapillary westerns and folding state-dependent immunoas-says. Based upon results from tauopathy cell culture model,the HGPS progeric mutation may be applied to animalstudies to augment the use of chronological ageing as aparameter in studies of Tau-mediated pathogenesis.

KEYWORDS: Tauopathy; laminopathy; progeria syn-drome; ageing; neurodegenerative disease

References

[1] Lee VM, Goedert M, Trojanowski JQ. Neurodegenerativetauopathies. Annu Rev Neurosci. 2001;24:1121–59.

PRION 31

[2] Nath U, Ben-Shlomo Y, Thomson RG, et al. The pre-valence of progressive supranuclear palsy (Steele-Richardson-Olszewski syndrome) in the UK. Brain2001;124(Pt 7):1438–49.

[3] Plassman BL, Langa KM, Fisher GG, et al. Prevalenceof dementia in the United States: the aging, demo-graphics, and memory study. Neuroepidemiology.2007;29:125–132.

[4] Onyike CU, Diehl-Schmid J. The epidemiology of fron-totemporal dementia. Int Rev Psychiatry. 2013;25(2):130–137.

[5] Mietelska-Porowska A, Wasik U, Goras M, et al. Tauprotein modifications and interactions: their role infunction and dysfunction. Int. J. Mol. Sci.2014;15:4671–4713.

[6] Frost, et al. Alzheimer’s disease: an acquired neurode-generative laminopathy. Nucleus. 2016;7(3):275–283.

[7] Frost, Bardai FH, Feany MB, et al. Lamin dysfunctionmediates neurodegeneration in tauopathies. CurrentBiol. 2016;26:129–136.

[8] Pollex RL, Hegele RA. Hutchinson-Gilford progeriasyndrome. Clin Genet. 2004;66(5):375–81.

[9] Schreiber KH, Kennedy, BK. When lamins go bad:nuclear structure and disease. Cell 2013. 152(6):1365–75.

[10] Jung HJ, Coffinier C, Choe Y, et al. Regulation ofprelamin A but not lamin C by miR-9, a brain-specificmicroRNA. Proc. Natl Acad. Sci. 2012;109:E423–E431.

[11] Yang SH, Procaccia S, Jung H-J. et al. Mice that expressfarnesylated versions of prelamin A in neurons developachalasia. Human Mol. Genet. 2015;24(10):2826–2840.

[12] Miller JD, Ganat YM, Kishinevsky S, et al. HumaniPSC-based modeling of late-onset disease via pro-gerin-induced aging. Cell Stem Cell. 2013;13:691–705.

[13] Sanders DW, Kaufman SK, DeVos SL, et al. Distincttau prion strains propagate in cells and mice and definedifferent tauopathies. Neuron 2014;82:1271–1288.

65. vCJD strain is consistent in individuals of twoPRNP codon 129 genotypes

Abigail B. Diacka, Aileen Boylea, Chris Plinstona, DianeRitchieb, Emma Hunta, Matthew Bishopb, Robert G.Willb and Jean C. Mansonc

aThe Roslin Institute and R(D)SVS, University of Edinburgh, EasterBush, UK; bNational CJD Research & Surveillance Unit, Center forClinical Brain Sciences, University of Edinburgh, Edinburgh, UK;cUniversity of Edinburgh, Edinburgh, UK

CONTACT Abigail B. Diack [email protected]

ABSTRACT

Background/Introduction: The identification of a clin-ical case of vCJD in a PRNP 129 heterozygous indivi-dual in 2016 raised concerns over whether 129MVindividuals can transmit the vCJD infection andwhether the 129MV genotype could give rise to a

novel strain of vCJD; both of which have significantpublic health concerns. To date, of the 178 definite/probable cases of vCJD, only one has been of the129MV genotype however evidence of abnormal PrPhas been observed in two asymptomatic cases of vCJD.We have now undertaken the strain characterization ofboth a clinical case of vCJD and that of an asympto-matic case of vCJD in 129MV individuals.Methods: CNS material from a 129MV clinical case ofvCJD [1] and spleen material from an asymptomatic129MV individual [2] were inoculated into panels ofwildtype mice. Serial passage of the spleen material fromthe asymptomatic 129MV individual was then performedby inoculating brain homogenate from a TSE positivemouse from each mouse strain into the wildtype mousepanel. This was then repeated for a secondmouse passage.A combination of clinical signs, neuropathology (TSEvacuolation and PrP deposition) and biochemical analysiswere carried out for each transmission and comparedagainst 129MM and BSE transmission data.Results: Primary passage of the clinical case of 129MVvCJD (CNS) in wildtype mice gave rise to pathologi-cally confirmed (TSE vacuolation and PrP deposition)clinical disease. Differences in transmission data wereobserved at the primary and first subpassage of theasymptomatic 129MV case (spleen) compared to129MM transmissions. These differences included lowattack rates with no or little TSE vacuolation at primarypassage and changes in incubation periods and rank-ings. These differences were resolved upon further pas-sage giving rise to strain characteristics consistent with129MM vCJD and BSE transmission data.Conclusions: These studies comprise the first strain char-acterization of vCJD in 129MV individuals. Upon pri-mary passage of the clinical case of 129MV vCJDtransmission characteristics resemble that of the129MMvCJD and BSE strain. While some differences wereobserved at primary and subpassage of the asymptomaticcase, these resolved by the second subpassage and straincharacteristics are totally consistent with 129MM vCJD.We demonstrate that vCJD strain properties are notaffected by transmission through individuals with the129MV genotype and thus no alteration in virulenceshould be associated with different host genotype.

References

[1] Mok T, Jaunmuktane Z, Joiner S, et al. VariantCreutzfeldt-Jakob disease in a patient with heterozyg-osity at PRNP codon 129. N Engl J Med. 2017;376(3):292–4.

32 ABSTRACT

[2] Peden AH, Head MW, Ritchie DL, et al. MokTPreclinical vCJD after blood transfusion in a PRNPcodon 129 heterozygous patient. Lancet. 2004;364(9433):527–9.

66. Scrapie strain typing of brain and lymph node-derived isolates in ovinized models reveals mixtureof substrains with distinct pathobiologicalproperties

Tomás Barrioa, Hicham Filalia, Alicia Oteroa, JessicaSheleby-Elíasa, Belén Marína, Enric Vidalb, Juan MaríaTorresc, Martin Groschupd, Olivier Andréolettie, JuanJosé Badiolaa and Rosa Boleaa

aCentro de Encefalopatías y Enfermedades Transmisibles Emergentes,Facultad de Veterinaria, Instituto Agroalimentario de Aragón – IA2,Universidad de Zaragoza – CITA, Zaragoza, Spain; bPriocat Laboratory,Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, UniversitatAutònoma de Barcelona (UAB), Barcelona, Spain; cCentro deInvestigación en Sanidad Animal, CISA-INIA, Madrid, Spain; dInstituteof Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institute,Greifswald-Isle of Riems, Germany; eUMR INRA ENVT 1225- IHAP, ÉcoleNationale Vétérinaire de Toulouse, Toulouse, France

CONTACT Rosa Bolea [email protected]

ABSTRACT

Phenotypic variability has been observed in severalprion diseases, including human TSEs and scrapie ofsheep of goats. This heterogeneity has been associatedwith distinct prion strains. The existence of strains inprion biology can be accommodated within the pro-tein-only hypothesis by supporting the view that severalconformational variants of PrPSc exist, which encodedistinct pathobiological properties. Within this frame-work, the conformational selection model proposes thata given amino acid sequence for PrPC allows a limitedportfolio of PrPSc conformations, such that the degreeof overlapping between host and donor Prnp sequencesdetermines the capability of the isolate to cause disease.

Transmission barrier occurs upon heterologous inter-action between incompatible PrP sequences, giving riseto a variety of new prion conformers. Therefore, trans-genic models expressing homologous PrPC are crucial tofaithfully study the actual variety of prion strains.Ovinized models show enhanced susceptibility to infec-tion with scrapie prions and have been employed tocharacterize strains in natural sheep isolates.

In the present study, we used the transgenic murinelines TgShp and Tg338, which express ovine PrPC ARQand VRQ, respectively, to bioassay 22 sheep scrapie isolatesfrom distinct outbreaks within the Spain-France-Andorratransboundary territory. Animals were intracerebrallyinoculated and survival periods, lesion profiles, PrPSc dis-tribution, and glycosylation patterns were studied.

In all cases but one, Western blot from sheep tissuesdisclosed glycosylation patterns compatible with a lowmolecular weight (Mw) scrapie strain (~19 kDa). OnlyInoculum 8L showed PrPres of higher Mw (~21 kDa).

On bioassay, all inocula caused on second-passageTgShp mice similar survival periods together with highattack rates. While most TgShp mice accumulated 19-kDaPrPres in their spinal cords, a number of isolates (includinginoculum 8L) triggered deposition of the 21-kDa isoform.

In Tg338 mice, a majority of isolates induced survivaltimes similar to those seen in TgShp, together with highattack rates and 19-kDa PrPres in the spinal cord. However,a group of isolates showed different features consisting invery long survival periods with low attack rates and pre-sence of 21-kDa PrPres. Additionally, these animals showedlower vacuolization scores in all evaluated brain areas andoccasional presence of amyloid plaques.

These results suggest that some scrapie isolates containmixtures of substrains that are resolved distinctly in dif-ferent transgenic lines. A number of isolates seemed tocomprise a major 19-kDa component and a minor 21-kDa component that is specifically amplified by theTgShp, but not the Tg338 line. In contrast, some otherisolates contained low titers of a 21-kDa, low pathogeni-city isoform that seemed to interfere with the propagationof themajor 19-kDa substrain exclusively in Tg338 thanksto its amyloidogenic properties, that sequester neurotoxicPrP monomers delaying the onset of clinical signs andrestraining the development of neuropathology.

67. Diagnostic accuracy of cerebrospinal fluid RT-QuIC in cases of suspected prion disease and thepotential utility of using RT-QuIC for public healthsurveillance

Daniel D. Rhoadsa,b, Aleksandra Wronac, Aaron Foutza,Curtis Tatsuokaa,d, Janis Blevinsa, Ryan A. Maddoxe,Ermias D. Belaye, Lawrence B. Schonbergere, Mark L.Cohena,b,f and Brian S. Applebya,b,f,g

aNational Prion Disease Pathology Surveillance Center, CaseWestern Reserve University, Cleveland, OH, USA; bDepartment ofPathology, Case Western Reserve University, Cleveland, OH, USA;cYale University, New Haven, CT, USA; dDepartment of Biostatistics,Case Western Reserve University, Cleveland, OH, USA; eCenters forDisease Control and Prevention, Atlanta, GA, USA; fDepartment ofNeurology, Case Western Reserve University, Cleveland, OH, USA;gDepartment of Psychiatry, Case Western Reserve University,Cleveland, OH, USA

CONTACT Brian S. Appleby [email protected]

ABSTRACT

Background: The clinical heterogeneity, potentialmimickers, and rapid progression make prion diseases

PRION 33

difficult to diagnose. The only way to definitely diagnoseprion disease is via neuropathologic examination, butreliable antemortem diagnoses can be made, especiallywith the advent of real time quaking induced conversion(RT-QuIC) to detect prions. Individuals with a neurop-sychiatric disorder and positive RT-QuIC are consideredto have probable sporadic Creutzfeldt-Jakob disease(sCJD) using current diagnostic criteria.Materials and Methods: Demographic and laboratorydata were examined for cerebrospinal fluid (CSF) speci-mens submitted to the National Prion Disease PathologySurveillance Center (NPDPSC) from May 2015 to April2018. In cases with multiple specimen submission, onlythe first specimen received was included in these analyses.Second generation RT-QuIC using truncated hamsterprion protein substrate was performed on all specimens.Neuropathologic examination at autopsy was used as thegold standard comparator. Autopsy is biased in favour ofcases where the diagnosis is uncertain at death, namelywhen false positive or false negative RT-QuIC results aresuspected. Autopsy tissue received through October 2018was included in the analyses.

Results: The NPDPSC received CSF specimens from10,498 living patients with suspected prion disease, and567 (5.4%) subsequently underwent neuropathologicexamination by the NPDSPC. Overall, 1,103 (10.5%) CSFspecimens submitted to the NPDPSC were RT-QuIC posi-tive. At autopsy, 497 (87.7%) cases were positive for priondisease, including 437 (77.4%)with definitive sCJD.OverallCSF RT-QuIC diagnostic sensitivity was 90.3% (449/497)and specificity was 98.5% (69/70). Sensitivity was highest insCJD (92.9%; 406/437) compared to genetic prion diseases(82%; 31/38). RT-QuICwas negative in all cases of sporadicfatal insomnia (sFI, n = 5), sCJD VV1 (n = 3), and fatalfamilial insomnia (FFI, n = 4). Younger subjects were morelikely to have false negative RT-QuIC results. One falsepositive RT-QuIC test occurred in a case of multifactorialdementia (Alzheimer’s disease and vascular).

Conclusions: We present analyses of a large cohort ofCSF specimens and autopsy tissue submitted to theNPDPSC that includes high variability in demo-graphics and prion disease subtypes. RT-QuICdemonstrated its highest sensitivity when testing CSFfrom older individuals and individuals with commonforms of sCJD. RT-QuIC demonstrated poor sensitiv-ity in detecting some uncommon prion disease sub-types (sFI, sCJD VV1, and FFI). The high specificityof RT-QuIC suggests that counting individuals whoare positive for RT-QuIC in public health surveillancecase numbers of probable/definitive prion disease, andantemortem CSF testing may foster improved priondisease surveillance.

68. Bioassays reveal Aβ and tau prions declinewith longevity in Alzheimer’s disease

Carlo Condelloa,b, Atsushi Aoyagia,c, Jan Stöhra,b,d,Weizhou Yuea, Brianna M. Riveraa, Joanne C. Leea,Amanda L. Woermana,b, Glenda Hallidaye, Sjoerd vanDuinenf, Martin Ingelssong, Lars Lannfeltg, CarolineGraffh,i, Thomas D. Birdj,k, C. Dirk Keenel, William W.Seeleyb,m, William F. DeGradoa,n and Stanley B.Prusinera,b,o

aInstitute for Neurodegenerative Diseases, Weill Institute forNeurosciences, University of California, San Francisco, CA, USA;bDepartment of Neurology, Weill Institute for Neurosciences,University of California, San Francisco, CA, USA; cDaiichi SankyoCo., Ltd., Tokyo, Japan; dAC Immune SA, Lausanne, Switzerland;eSchool of Medical Science, University of New South Wales, Sydney,Australia; fLeiden University Medical Center, Leiden, TheNetherlands; gDepartment of Public Health and Caring Services/Geriatrics, Uppsala University, Uppsala, Sweden; hDepartment ofNeurobiology, Care Sciences and Society, Karolinska Institute,Huddinge, Sweden; iDepartment of Geriatric Medicine, KarolinskaUniversity Hospital, Stockholm, Sweden; jDepartment of Medicine,Division of Medical Genetics, University of Washington, Seattle, WA,USA; kDepartment of Neurology, University of Washington, Seattle,WA, USA; lDepartment of Neuropathology, University ofWashington School of Medicine, Seattle, WA, USA; mDepartmentof Pathology, University of California, San Francisco, CA, USA;nDepartment of Pharmaceutical Chemistry, University of California,San Francisco, CA, USA; °Department of Biochemistry andBiophysics, University of California, San Francisco, CA, USA.

CONTACT Carlo Condello [email protected]

ABSTRACT

We present evidence that both the Aβ and tau proteinsmisfold into prions leading to Alzheimer’s disease (AD),which is a sporadic or genetic dementing disorder but notcommunicable or contagious. Since the progression ofAD correlates poorly with insoluble Aβ in the centralnervous system (CNS), it was difficult to distinguishbetween inert amyloids and Aβ prions. To measure theprogression of AD, we devised exquisitely selective andsensitive bioassays to measure the abundance of isoform-specific Aβ prions in human CNS samples from patientswith AD or other neurodegenerative diseases. Althoughbrains from all human AD patients had measurable Aβprions at death, the oldest individuals had lower Aβ prionlevels. Long-lived individuals had low tau prion levels thatcorrelated with phosphorylated tau. Surprisingly, thelongevity-dependent decrease in tau prions occurred inspite of increasing amounts of total insoluble tau. Whencorrected for the abundance of insoluble tau, the tauprions decreased exponentially with respect to the age ofdeath with a half-time of approximately one decade, andthis correlated with the abundance of phosphorylated tau.Our findings with tau prions were not only unexpectedbut also counterintuitive; thus, tau phosphorylation andtau prion activity decreased exponentially with longevity

34 ABSTRACT

in patients with AD ranging from ages 37 to 99 years. Ourfindings demonstrate an inverse correlation betweenlongevity in AD patients and the abundance of neurotoxictau prions. Moreover, our discovery may have profoundimplications for the selection of phenotypically distinctpatient populations and development of diagnostics andeffective therapeutics for AD.

69. Why is there a decline in the incidence ofsporadic Creutzfeldt Jakob Disease in the olderadult population?

Investigating mixed pathologies in sporadicCreutzfeldt Jakob Disease

Jane Lumsden, Bob Will, Colin Smith and Suvankar Pal

National CJD Research and Surveillance Unit, University of Edinburgh

ABSTRACT

Background: Sporadic Creutzfeldt Jakob Disease (sCJD) isa rapidly progressive and fatal neurodegenerative disorderand the commonest occurring form of human prion dis-ease. Age-specific mortality rates for sCJD have increasedup to ages 65–79 years over the past four decades in theUnited Kingdom. Of considerable interest is an apparentreduced incidence at ages of 80 and over. A similar phe-nomenon has been observed in other European countries.This reduced incidence could be due to an under ascertain-ment in the most elderly. It has also been recently hypothe-sized that the apparent decline in incidence of sCJD inolder adults could be due to the inhibitory effects of theAlzheimer’s disease associated amyloid β-protein(A β) onprion propagation. Aβ deposition is common in advancedage, even in cognitively asymptomatic individuals, and arecent study suggested that soluble aggregates of Aβ cancompete with prions for binding to misfolded cellularprion protein. This study raised the possibility that pre-sence of Alzheimer’s disease pathology may be protectiveagainst CJD. Another recent neuropathological studyinvestigating 30 cases with confirmed sCJD reported thatco-morbid presence of abnormal proteins associated withother neurodegenerative disorders including Alzheimer’sdisease, amyloid angiopathy, Lewy body dementia, argyr-ophilic grain disease, and progressive supranuclear palsywas found to be significantly associated with prolongedsurvival. This led authors to suggest that presence ofdual/multiple neurodegenerative pathologies may be con-tributing to this phenomenon. Furthermore, additionalwork has demonstrated that sCJD cases with abundantdeposits of Aβ present at a higher median age of diseaseonset and have longer disease duration. This project pro-posal identifies aims and hypotheses for this ongoingproject.

Aims:

(1) To review UK postmortem reports of patientswith confirmed sCJD to establish presence ofβ-amyloid, neurofibrillary tangles, neuritic pla-ques, α synuclein, amyloid angiopathy, andrecord Thal phase and Braak stage.

(2) To review clinical details of these cases includ-ing clinical co-morbidities, age at onset ofsCJD, presenting symptoms, disease duration,codon 129 status, MRI findings, 14–3-3, andRT-QuIC results

(3) To compare clinical features and investigationresults of cases with dual/multiple co-patholo-gies with cases with no known co-pathology.

Hypotheses:(1) Cases with neuropatholgical evidence of dual

or multiple pathologies will present differentlyto cases with isolated sCJD pathology.

(2) MRI scans will show focal atrophy, or vascularburden which may confound the typical corti-cal and basal ganglia changes seen in sCJD.

(3) CSF 14–3-3 and RT-QuIC results will differ.Specifically, 14–3-3 will be negative in cases ofdual/multiple pathologies.

(4) Individual co-pathologies will influence theclinical features and investigation results in adifferential manner.

(5) Cases with significant Lewy body deposition willlikely test positive for alpha-synuclein in CSF.

70. A histidine swap in the prion protein’s CopperCenter Drives a neuroprotective cis-interaction

Kevin Schillinga, Lizhi Taob, Bei Wuc, GlennMillhausera, David Brittb and David Harrisc

aDepartment of Chemistry & Biochemistry, UC Santa Cruz, SantaCruz CA, USA; bDepartment of Chemistry, UC Davis, Davis, CA, USA;cDepartment of Biochemistry, Boston University School of Medicine,Boston, MA, USA

CONTACT Kevin Schilling [email protected]

ABSTRACT

The cellular prion protein (PrPC) is comprised of twodomains – a globular C- terminal domain and an unstruc-tured N-terminal domain. Recently, copper has beenobserved to drive tertiary structure in PrPC, inducing acis-interaction between the two domains of the protein.1–3 The location of this interaction on the C-terminus over-lapswith the sites of PrPSc docking, toxic antibody docking,and pathogenicmutations. Combined with recent evidencethat the N-terminus is a toxic effector regulated by the C-

PRION 35

terminus,4,5 there is a significant possibility that this cis-interaction serves a protective role, and that the disruptionof this interactionbyPrPSc oligomers is the cause of toxicityin prion disease. We demonstrate here that two highlyconserved histidines in the C-terminal domain of PrPCare essential for the protein’s cis-interaction, which helpsto protect against neurotoxicity carried out by its N-termi-nus. We show that mutation of these histidines drasticallyweakens the cis-interaction and biases cultured cellstowards toxic events – the first C-terminal mutation to doso. Mechanistically, we propose an equatorial swap – acopper bound histidine from theN-terminus of the proteinis replaced by a histidine from its C-terminus, forming atether that holds the two domains together. We also findthat extra N-terminal histidines in pathological familialmutations involving octarepeat expansions inhibit thisinteraction by stealing copper from the C-terminus, sug-gesting a mechanism for the toxicity of these mutants. Webelieve we have found the structural basis for PrPC’s C-terminal regulation of its N-terminus, and that thismechanism is responsible for preventing neurotoxicity inthe properly functioning form of the protein.

References

[1] Thakur, et al. JBC 2011; 286:38,533–38,545.[2] Spevacek, et al. Structure 2013; 21:236–246.[3] Evans, et al. Structure 2016; 24:1057–10674.[4] Sonati, et al. Nature 2013; 501:102–106.[5] Wu, et al. eLife 2017. doi:10.7554/eLife.23,473.001

71. Challenging assumptions: an empiricalanalysis of hunter response to chronic wastingdisease in Alberta

John K. Pattison-Williamsa, Lusi Xiea, Vic (W.L.)Adamowicza, Margo Pybusb and Anne Hubbsb

aDepartment of Resource Economics and Environmental Sociology,University of Alberta, Edmonton, Alberta, Canada; bAlbertaEnvironment and Parks, Edmonton, Alberta, Canada

CONTACT John K. Pattison-Williams [email protected]

ABSTRACT

Background: Chronic Wasting Disease (CWD) has fun-damentally impacted wildlife management in NorthAmerica. An integral partner in CWD managementstrategies is the hunting community, whose membershipcontributes in surveillance, harvest, and policy develop-ment. A conventional assumption has been that hunterswill avoid or reduce hunting in areas where CWD isidentified as high prevalence due to potential humanhealth risks [1], requiring policy incentives to promote

continued or increased hunting in these areas. However,this assumption has recently been challenged [2].Methods: Using mule deer hunter draw information andCWD surveillance data from the Alberta Ministry ofEnvironment and Parks (AEP), we empirically explore theresponse of mule deer hunters to CWDusing license appli-cation trends in wildlifemanagement units (WMUs) whereCWD has been positively detected. Using a fixed effects(FE) approach, we model the relationship between totalresident draw applications and covariates of CWD headcounts, quotas, draw success rates between 2005 and 2016in 39 WMUs.Results: Our model indicates that mule deer hunters inAlberta are continuing to hunt in WMUs with high CWDprevalence and no significant relationship exists betweenCWD count and draw applications. These results suggestthat (i) hunter decisions regarding license applications arenot significantly affected by CWD presence or prevalence,or (ii) hunters perceive CWD in a WMU as a factor thatdoes not detract from, and may be attractive for huntingopportunities.Conclusions: Our results have important policy implica-tions for CWD management. First, it challenges theassumption that hunters are avoiding CWD zones.Second, it suggests that alternative hunter-based strategies– such as expanded hunting seasons, increased tags orreplacement tags – may be more effective and less costlythan previously thought to manage the disease. One caveatis that our analysis is based on analyses of aggregatedemand for hunting licenses at the WMU level, ratherthan individual hunter demands over time.

KEYWORDS: Chronic wasting disease; recreationalhunting; cervids; alberta; mule deer; wildlifemanagement

References

[1] Cooney, et al. Hum Dimens Wildl 2010; 15: 194–207.[2] Haus, et al. Wildl Soc Bull 2017; 41:294–300.

72. miR-146a regulates cellular and diseaseassociated prion protein isoforms during prionpropagation

Wenting Zhaoa,b, Cathryn Ugaldea, Steven Collinsc,Vicki Lawsond, Lesley Chenga, Shayne Bellinghamb

and Andrew Hilla

aDepartment of Biochemistry and Genetics, La Trobe Institute forMolecular Science, La Trobe University, VIC, Australia; bDepartment ofBiochemistry and Molecular Biology, The University of Melbourne, VIC,Australia; cFlorey Institute of Neuroscience and Mental Health, TheUniversity of Melbourne, VIC, Australia; dDepartment of Microbiology

36 ABSTRACT

and Immunology, The Peter Doherty Institute for Infection andImmunity, The University of Melbourne, VIC, Australia.

CONTACT Wenting Zhao [email protected]

ABSTRACT

Prion diseases are transmissible neurodegenerativedisorders characterized by the structural transforma-tion of the cellular prion protein (PrPC) to the priondisease associated isoform (PrPSc). MicroRNAs(miRNAs) are a class of small non-coding RNAsthat regulate gene or protein expression by targetingmRNAs and triggering either translational repressionor mRNA degradation. Distinct miRNA signatureshave been detected in prion disease models andpatients, serving as potential disease diagnostic bio-markers. In this study, increased expression levels ofmiR-146a, an anti-inflammatory miRNA known to bedysregulated in prion diseases, were detected in fourprion disease models: in prion strain M1000 infectedneuronal N2a cells; at day 42 and day 70 of M1000infected organotypic brain slices; at week 17 andterminal stages of M1000 infected mouse brains;and in the post-mortem brain tissue samples fromsporadic Creutzfeldt-Jakob disease (sCJD) patients.CRISPR/Cas9 mediated miRNA editing and prioninfection were further used to examine the effects ofmiR-146a gain/loss-of-function on prion proteins incell culture and organotypic brain slice culture mod-els. miR-146a was shown to increase PrPC levelswhen overexpressed or downregulated in N2a cells.The prion protein encoding gene (Prnp) was shownto be targeted by miR-146a in the 3’ untranslatedregion (3’ UTR). miR-146a overexpression decreasedthe amount of PrPSc in M1000 infected N2a cells,while M1000 infected organotypic brain slices frommiR-146a knockout mice exhibited increased PrPSc

levels compared to the wildtype controls. Overall,the protective role of miR-146a was revealed inprion diseases by regulating cellular and disease asso-ciated prion proteins, suggesting its therapeuticpotential.

73. Transgenic overexpression of the PrP N1fragment does not protect against prion disease orAβ toxicity due to impaired ER translocation

Behnam Mohammadia, Luise Linsenmeiera, MohsinShafiqa, Berta Puiga, Giovanna Galliciottia, JörgTatzeltb, Markus Glatzela* andHermannC. Altmeppena*aInstitute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; bBiochemistry ofNeurodegenerative Diseases, Institute of Biochemistry andPathobiochemistry, Ruhr University Bochum, Bochum, Germany

CONTACT Behnam Mohammadi [email protected]

*These authors contributed equally to this work.

ABSTRACT

The highly conserved and constitutively active endogen-ous α-cleavage of the prion protein has so far mostly beenlinked with protective effects. As a soluble factor, thereleased unstructured N1 fragment acts beneficial in sev-eral ways as it, for instance, reduces hypoxia-inducedneuronal damage and may be involved in myelin main-tenance and other aspects of intercellular communication.With regard to neurodegenerative diseases, numerousstudies have shown that N1 is able to block toxic oligo-mers, such as Aβ in Alzheimer`s disease, and interfereswith their synaptic impairment and neurotoxicity.

For prion diseases, however, insight into a poten-tially protective role of N1 by similarly neutralizingPrPSc oligomers and interfering with prion conversionis lacking to date. Since the responsible protease for theα-cleavage has not been identified unequivocally, phar-macological targeting of this entity is not possible todate. We therefore decided to directly address this issuein vivo by generating transgenic mice (tgN1) overex-pressing N1 on a wild-type background and challengingthem with RML prions. Despite moderate differences inPrPSc formation and p38 activation, incubation time,and disease duration was similar between tgN1 miceand wild-type littermate controls. Biochemical andmorphological assessment of brain samples, primaryneurons, and cell culture models then revealed, thatour direct ‘protective’ strategy failed due to cytosolicaccumulation and lack of secretion of the transgenicN1. Nevertheless, our study provides the first in vivoproof of the recently described impaired translocationof intrinsically disordered peptides into the endoplas-mic reticulum. Moreover, it demonstrates effects ofcytosolic accumulation of N1 with uncleaved signalpeptide, addresses proteasomal degradation, questionsa general relevance of cytosolic prions in prion diseases,and highlights important aspects to be considered wheninvestigating the α-cleavage of PrPC or devising N1-based therapeutic approaches.

74. Characterization of gene expression profiles inbrains of mice infected with typical and atypicalBSEsTakeshi Yamasakia, Akio Suzukia, Rie Hasebeb, YuichiMatsuurac, Kohtaro Miyazawac, Yoshifumi Iwamaruc

and Motohiro Horiuchia

aLaboratory of Veterinary Hygiene, Faculty of Veterinary Medicine,Hokkaido University, Japan; bInstitute for Genetic Medicine,

PRION 37

Hokkaido University, Japan; cNational Institute of Animal Health,National Agriculture and Food Research Organization (NARO),Japan

CONTACT Takeshi Yamasaki [email protected]

ABSTRACT

Transgenic mice overexpressing bovine PrP (TgBoPrP)infected with typical BSE (C-BSE), atypical L-type BSE,andH-type BSE (L-BSE andH-BSE) show different clinicaland neuropathological characteristics. To understand hostresponses that contribute to the differences in the pheno-types, we analysed gene expression profiles in brains ofTgBoPrP mice infected with C-BSE, L-BSE, and H-BSE.We performed a transcriptome analysis by RNA-sequen-cing on brainstems of the mice at pre-clinical, clinical, andterminal stages of infection and those of age-matched un-infectedmice. First, we performed hierarchical clustering ofsamples based on gene expression data using 16,894 genesthat code proteins. The samples frommice infected with L-BSE at 198 days post intracerebral inoculation (dpi), C-BSEat 239 and 253 dpi, andH-BSE at 291, 319 and 362 dpi wereclearly separated from cluster containing un-infected sam-ples, suggesting that host responses accompanied withmarked transcriptomic changes initiated by 198, 239, and291 dpi in L-BSE, C-BSE and H-BSE infected mice, respec-tively. Next, we identified 3,219 differentially expressedgenes (DEGs) that showed over 2-fold change in expressionlevel between infected and un-infected mice. By applyingK-means clustering to the DEGs, we found four character-istic gene clusters that contain genes of which expressionwas upregulated by infection. Three clusters contained 338,517 and 294 upregulated genes unique to C-BSE, L-BSE,and H-BSE, respectively, and the other cluster contained251 upregulated genes common to all BSEs. Gene ontology(GO) analysis based on Metascape database revealed thatgenes related to immune responses were highly enriched inthe upregulated genes common to all BSEs. Althoughenrichment scores were not so high, the upregulatedgenes unique to each BSE showed a significant enrichmentof GO terms related to regulation of immune system. Thesedata might suggest the presence of immune responses bothcommon and unique to C-BSE, L-BSE, and H-BSEinfection.

75. Mortality surveillance of individuals potentiallyexposed to chronic wasting disease

Ryan A. Maddoxa, Rachel F. Klosb, Suzanne N. Gibbons-Burgenerb, Bobbi L. Bryanta, Joseph Y. Abramsa, Brian S.Applebyc, Lawrence B. Schonbergera and Ermias D.Belaya

aNational Center for Emerging and Zoonotic Infectious Diseases, Centersfor Disease Control and Prevention (CDC), Atlanta, GA, USA; bWisconsinDepartment of Health Services, Division of Public Health, Madison, WI,USA; cNational Prion Disease Pathology Surveillance Center (NPDPSC),Case Western Reserve University, Cleveland, OH, USA

CONTACT Ryan A. Maddox [email protected]

ABSTRACT

Introduction: Chronic wasting disease (CWD) is a priondisease of cervids. It is unknown whether CWD prionscan infect people; if so, transmission would most likelyoccur through consumption of meat from infected ani-mals. Since 2003, Wisconsin Department of HealthServices, Division of Public Health (WDHS) personnelhave maintained a database consisting of informationcollected from hunters who reported eating, or an inten-tion to eat, venison from cervids positive for CWD. Thisdata source makes it possible to evaluate causes of mor-tality in individuals potentially exposed to CWD.Methods: The WDHS database contains the name, dateof birth, when available, year of CWD-positive deerharvest, and city and state of residence for each poten-tially exposed individual. The database also includesinformation on how the deer was processed (self-pro-cessed or by a commercial operator) and when applic-able, names of others with whom the venison wasshared. Duplicate entries (i.e. those who consumed veni-son from CWD-positive deer in multiple hunt years) aredetermined by first name, last name, and date of birth.All names in the database are cross-checked with theNational Prion Disease Pathology Surveillance Center(NPDPSC) neuropathology database. Vital status ofindividuals with date of birth available will be trackedthrough the identification of possible matches in theNational Death Index (NDI) and the evaluation of cor-responding cause of death codes.Results: The database consists of 1561 records for huntyears 2003–2017. Of these, 613 records had accompany-ing date of birth; 14 entries were removed as duplicates, 1of whom had consumed venison from a CWD-positivedeer during three different hunt years, leaving 599 uniqueindividuals for pending submission to NDI. Of theseindividuals, 265 of 399 (66%) who ate venison from aCWD-positive deer and provided processing informationreported self-processing. No matches were found amongpersons in the database cross-checked with NPDPSCdata.Conclusion: Because of the robust data link between per-son and CWD-positive animal, reviewing the cause ofmortality in potentially exposed persons is possible; thoseindividuals who self-processed and consumed the meat are

38 ABSTRACT

likely the best source of information about the potential forzoonotic transmission. The expected long incubation per-iod, should transmission to humans occur, necessitatesmany years of vital status tracking.

76. Prion disease incidence, United States, 2003–2016

Ryan A. Maddoxa, Marissa K. Persona, Janis E. Blevinsb,Joseph Y. Abramsa, Bobbi L. Bryanta, Brian S. Applebyb,Lawrence B. Schonbergera and Ermias D. Belaya

aNational Center for Emerging and Zoonotic Infectious Diseases,Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA;bNational Prion Disease Pathology Surveillance Center (NPDPSC),Case Western Reserve University, Cleveland, OH, USA

CONTACT Ryan A. Maddox [email protected]

ABSTRACT

Introduction: Mortality data and the results of neuro-pathological and genetic testing are used to estimate theincidence of prion diseases in the United States.Methods: Prion disease decedents were identified fromrestricted-use U.S. national multiple cause-of-deathdata, via a data use agreement with the NationalCenter for Health Statistics, and from the NationalPrion Disease Pathology Surveillance Center(NPDPSC) database for 2003–2016. NPDPSC dece-dents with neuropathological or genetic test resultspositive for prion disease for whom no likely matchwas found in the multiple cause-of-death data wereadded as cases for incidence calculations; those withnegative neuropathology results but with cause-of-death data indicating prion disease were removed.Age-adjusted average annual incidence rates for thecombined data were calculated using the year 2000 asthe standard population.Results: A total of 5737 decedents were identified ashaving prion disease during 2003–2016 for an age-adjusted average annual incidence of 1.2 per millionpopulations. The age-adjusted annual incidence rangedfrom 1.0 per million in 2004 and 2006 to 1.4 per millionin 2013; there was an increasing trend in age-adjustedincidence over the entire period (p <0.0001) but nosignificant increase during the second half (2010–2016, p = 0.08). The age-adjusted incidence betweenmales and females (1.3 and 1.1 per million, respec-tively) differed significantly (p <0.0001). Twelve casesduring 2003–2016 were <30 years of age for an age-specific incidence of 6.9 per billion; only two of thesevery young cases were sporadic, with the rest beingfamilial (7), variant (2), or iatrogenic (1). The age-specific average annual incidence among those <55and ≥55 during the time period was 0.2 and 4.7 per

million, respectively; incidence among those ≥65 was5.9 per million.Conclusion: The incidence of prion diseases, which areinvariably fatal, can be estimated through analysis ofmortality data supplemented with the results of neuro-pathological and genetic testing. Incidence in theUnited States over the last 7 years appears to be rela-tively stable. Cases <30 years of age continue to be veryrare and usually indicate an exogenous source of infec-tion or the presence of a genetic mutation.

77. Assessing chronic wasting disease straindifferences in free-ranging cervids across theUnited States

Kaitlyn M. Wagnera, Caitlin Ott-Connb, Kelly Strakab,Bob Dittmarc, Jasmine Battend, Robyn Piercea,Mercedes Hennessya, Elizabeth Gordona, BrettIsraela, Jenn Ballarde and Mark D Zabela

aPrion Research Center at Colorado State University; bMichiganDepartment of Natural Resources; cTexas Parks and WildlifeDepartment; dMissouri Department of Conservation, 5. ArkansasGame and Fish Commission

CONTACT Kaitlyn M. Wagner [email protected]

ABSTRACT

Background/Introduction: Chronic wasting disease(CWD) is an invariably fatal prion disease affectingcaptive and free-ranging cervids, including white-taileddeer, mule deer, moose, elk, and reindeer. Since theinitial description of the disease in the 1960’s, CWD hasspread to 23 states, 3 Canadian Provinces, South Korea,Norway and, most recently, Finland. While some out-breaks of CWD were caused by transport of infectedanimals from endemic regions, the origin of CWD inother epizootics is unclear and has not been character-ized. Previous studies have shown that there are twodistinct strains of CWD. However, the continuousspread and the unclear origin of several outbreaks war-rant continued surveillance and further characteriza-tion of strain diversity.Materials and Methods: To address these knowledgegaps, we used biochemical tests to assess strain differ-ences between CWD outbreaks in Michigan, Texas,Missouri, and Colorado, USA. Brain or lymph nodesamples were homogenized and digested in 50 µg/mLproteinase K (PK). These samples were then run on aWestern blot to assess glycoform ratio and electro-phoretic mobility. Texas samples were digested in100 µg/mL PK. To assess conformational stability,brain or lymph node homogenates were incubated inincreasing concentrations of guanidine hydrochloridefrom 0 M to 4 M in 0.5 M increments. Samples were

PRION 39

then precipitated in methanol overnight, washed andPK digested in 50 µg/mL PK before slot blotting.Results: Our results have found significant differences inglycoform ratio between CWD from Michigan andColorado, but no differences were observed in conforma-tional stability assays. Interestingly, when testing our CWDisolates from Texas to analyse electrophoretic mobility andglycoform ratio, we found that these samples did notexhibit the characteristic band shift when treated withPK, but PK resistant material remained. Additionally,results from our conformational stability assay demon-strate a unique profile of these Texas isolates. Testing ofsamples from Missouri is currently underway.Conclusions: Thus far, our data indicate that there arestrain differences between CWD circulating in Michiganand CWD in Colorado and provide important insight intoCWD strain differences between two non-contiguous out-breaks. We have also identified a unique strain of CWD inTexas with biochemical strain properties not seen in any ofour other CWD isolates. These results highlight the impor-tance of continued surveillance to better understand thisdevastating disease. These results have important implica-tions for CWD emergence, evolution and our understand-ing of prion strain heterogeneity on the landscape.

78. Optogenetically stimulation of medial septum-diagonal band complex ameliorates the cognitivedysfunctions of APPNL-G-F, mouse model ofAlzheimer’s disease

Jogender Mehla, Surjeet Singh, Robert J. McDonaldand Majid H. Mohajerani

Canadian Centre for Behavioural Neuroscience, University ofLethbridge, Lethbridge, Alberta, Canada.

CONTACT Jogender Mehla [email protected]; [email protected]

ABSTRACT

Introduction: Cholinergic neuron degeneration in thebasal forebrain is known to be one of the initial patholo-gical hallmarks of Alzheimer’s disease (AD). Additionally,currently available drug therapy is also based on thecholinergic hypothesis which provides only symptomatictreatment. Therefore, considering the importance of cho-linergic system inAD, we planned this study to investigatethe effect of chronic optogenetic stimulation of medialseptum-diagonal band (MSDB) complex, a part of basalforebrain on the cognitive deficits and amyloid pathologyin knock-in mouse model of AD.Materials and Methods: In the present study, we usedmale APPNL-G-F mice. We used male C57BL/6J mice as anormal control. We injected channelrhodopsin virus and

implanted optrodes into MSDB complex of 2 months oldAPPNL-G-F mice. After one month of surgery, these micewere divided into two groups; one, where no optogeneticstimulation was done and second, in which MSDB stimu-lation was performed for 3 months using blue light(473 nm, 3–4 mW). After end of 3 months stimulation,Morris water maze (MWM) and fear conditioning testswere performed for the assessment of learning and mem-ory functions. Lastly, we perfused the mice and brains wereextracted for immunostaining.Results: The results show that optogenetically stimulationofMSDB improved the learning ability of APPNL-G-F micein MWM evidenced by significant decrease in escapelatency on day 8 in comparison with APPNL-G-F shamgroup. Furthermore, optogenetic stimulated mice spentsignificantly more time in target quadrant as compared toaverage of other quadrants. Nonetheless, optogeneticallystimulated mice also spent significantly more time intarget quadrant in comparison with APPNL-G-F shamgroup indicating an improvement in memory retention.APPNL-G-F mice also showed an improvement in memoryfunction in tone and contextual fear conditioning tests.The chronic optogeneticallly stimulation of MSDBreduced the amyloid burden in cortex and hippocampusof APPNL-G-F mice which may be responsible forimprovement in cognitive functions.Conclusion: The findings from the present study sup-port the potential of cholinergic hypothesis and opto-genetics for the treatment of AD.

79. The FXR1 protein is a functional amyloid ofmammalian brain

A. V. Sergeevaa, M. E. Velizhaninaa, J. V. Sopovaa,b, E. I.Koshelc, T. A. Belashovab, S. P. Zadorskya,b, V. A.Siniukovab and A. P. Galkina,b

aDepartment of Genetics and Biotechnology, St. Petersburg StateUniversity, Petersburg, Russian Federation; bVavilov Institute ofGeneral Genetics, St. Petersburg Branch, Russian Academy ofSciences, Petersburg, Russian Federation; cDepartment ofCytology and Histology, St. Petersburg State University, St.Petersburg, Russian Federation

CONTACT A. V. Sergeeva [email protected]

Amyloids are non-branching fibrils that are composed ofstacked monomers stabilized by intermolecular β-sheets.Our novel proteomic approach PSIA-LC-MALDI sepa-rates amyloids from the majority of other protein com-plexes of a non-amyloid nature. Being applied to R.norvegicus brain proteome, PSIA-LC-MALDI allowedidentification of several amyloid-like proteins formingSDS-resistant aggregates. FXR1 was among identifiedproteins. FXR1 binds and regulates translation of targetmRNAs and takes part in memory and emotions control.

40 ABSTRACT

We demonstrated that FXR1 forms amyloid conformersthat strongly co-localize with thioflavin S, bind RNA andprevent its RNAseA-mediated degradation in the cortexneurons of the rat brain. Furthermore, we showed that N-terminal region of the FXR1 protein underlies the abilityof full-length FXR1 to form SDS-resistant oligomers andinsoluble aggregates in the brains of young and healthy ratmales and forms amyloid fibrils in vitro. Moreover, invitro assay confirmed the ability of amyloid conformers ofthe N-terminal region of FXR1 to bind poly(A)-RNAextracted from the rat brain. To further evaluate theamyloidogenic properties of FXR1 N-terminal region weused bacterial-based C-DAG system. Not to our surprise,FXR1(1–380) forms extracellular fibrils in bacteria, thatcan be visualized by TEM and demonstrates apple-greenbirefringence when stained with Congo Red and viewedunder crossed polarizers. Specification of FXR1’s amyloi-dogenic region boundaries allowed us to shorten it from(aa 1–380) to (aa 1–270), as FXR1(1–270) demonstratessame amyloidogenic properties as FXR1(1–380) in C-DAG system. To shed the light on the possibility of theFXR1 protein being a functional amyloid of human brainas well, we used a human neuroblastoma cell line, IMR32,as a human brainmodel.We demonstrated that in humanneuroblastoma cells FXR1 forms insoluble aggregates ofhighmolecular mass the abundance of which is controlledin serum-dependent manner. Considering the fact thatserum starvation is one of the standard methods of indu-cing neuroblastoma cells differentiation, we speculate thatFXR1 oligomers and amyloid aggregates may play a rolein cell differentiation via its interaction with mRNAs ofcell cycle proteins, such as p21. Interestingly, the N-term-inal region of FXR1 is highly conserved across mammalsand contains identically arranged potentially amyloido-genic sequences in evolutionary distinct vertebrates.These data support our hypothesis that the ability ofFXR1 to function in amyloid form is conserved in evolu-tion. The reported study was funded by RFBR accordingto the research project 18–34-00420 and grant of SpbSUto Laboratory of Amyloid Biology.

80. Cellular prion protein promotes cell metabolicorientation towards the oxidative degradation ofglucose

H. Arnoulda, A. Baudrya, M. Pietria, A. M. Haeberléb,M. Laforgea, G. Berthoc, G. Schmitt-Ulmsd, Y. Baillyb,O. Kellermanna and B. Schneidera

aINSERM UMR 1124, Université Paris Descartes, Paris, France; bCNRSUPR 3212, Institut des Neurosciences Cellulaires et Intégratives,Strasbourg, France; cCNRS UMR 8601, Université Paris Descartes,Paris, France; dTanz Centre for Research in NeurodegenerativeDiseases, University of Toronto, Canada

CONTACT H. Arnould [email protected]

While the implication of cellular prion protein PrPC hasbeen widely documented in prion diseases, the role(s) ofthis ubiquitous protein still remain(s) elusive. To addressthis issue, PrPC was chronically silenced in the 1C11neuronal cell line, a neuronal stem cell endowed withthe capacity to differentiate into serotonergic or noradre-nergic neurons. A global analysis was performed to com-pare the proteome of PrPC-depleted 1C11 cells (referredto as PrPnull-1C11 cells) to that of parental PrPC-expres-sing 1C11 cells with the aim to identify cell function(s)deregulated by the absence of PrPC. Our proteomic ana-lysis reveals strong abnormalities in the expression level ofenzymes at play in the glucose energetic metabolism inPrPnull-1C11 cells with a reduced expression of mito-chondrial enzymes in favour of the enzymes involved inthe glycolytic-fermentative pathway. Surprisingly, func-tional experiments by NMR and Seahorse approaches toprobe metabolic fluxes in cells indicate reduced glycolyticand lactic fermentative fluxes as well as decreased mito-chondria oxygen consumption in PrPnull-1C11 cellswhen grown in a medium containing glucose. Globalreduction of glucose oxidative degradation in PrPC-depleted cells is associated with down-expression of theglucose transporter GLUT3 and down-regulation of pyr-uvate dehydrogenase (PDHA1) activity that normallypromotes the entry of pyruvate into mitochondria andtransformation into acetyl-CoA. Finally, we show a meta-bolic shift in PrPnull-1C11 cells with a preferred orienta-tion of the energetic metabolism towards fatty aciddegradation by the β-oxidation pathway to maintainATP concentration at high levels. The flip side of suchconversion in the energetic pathways is the onset of oxi-dative stress conditions in PrPC-depleted cells. For thefirst time, our work provides evidence that PrPC plays akey role in the energetic metabolism by potentiating glu-cose oxidative degradation through the glycolysis- mito-chondria pathway. As corruption of PrPC function(s) bypathogenic prions is at the root of prion diseases, anyperturbation of PrPC control of glucose degradation inprion-infected cells may contribute to neurodegeneration.

KEYWORDS: Proteomics; PrPC function; glucose andfatty acid energetic metabolisms; oxidative stress

81. Detection of neuronal dysfunction associatedwith genetic prion disease in human cerebralorganoids

Simote T. Foliakia, Bradley R. Grovemana, AleksandarR. Wooda, Jue Yuanb, Leslie Coopermanb, Paul Tesarb,Wen-Quan Zoub and Cathryn L. Haigha

PRION 41

aNational Institute of Allergy and Infectious Diseases, NIH; bCaseWestern Reserve University School of Medicine

CONTACT Simote T. Foliaki [email protected]

ABSTRACT

Introduction: One of the fundamental problems ofresearching human prion diseases is access to livehuman brain tissue. Utilizing stem cell technology toproduce human cerebral organoids (COs), which cur-rently represent the nearest model to structured humanbrain tissue, offers a new avenue of investigation [1].These organoids are self-organizing, structured regionsanalogous to human brain tissue that mature to containmultiple neuronal populations as well as astrocytes andoligodendrocytes [1, 2]. Importantly, these COs could begenerated from donors who are carriers of mutationsassociated with familial prion diseases such as theE200K mutation. Herein we aimed to investigate theinfluence of E200K mutation that pre-dispose individualsto prion disease on neuronal electrical signalling.Methods: COs have been generated from donors withE200K mutations (pre-disposed) and without (non-predisposed) mutations within the prion gene usingthe protocol described by Lancaster and Knoblick [1].To measure the ability of these organoids to evokespontaneous neuronal electrical signalling, they weresliced into 300 µm thick slices and mounted ontomulti-electrode arrays (MEAs). MEAs are embeddedwith micro-electrodes to measure extracellular neu-ronal electrical spike (action potentials) and networkcommunication throughout individual organoid.During the recording, the organoid slices were pro-vided with adequate nutrients and oxygen throughcontinuous superfusion with oxygenated artificialcerebrospinal fluid. To induce electrical signallingassociated with cognition and behaviour, we exposedthe slices to known stimulants and depressants ofneuronal activity to determine their ability to expressfunctions associated with memory formation andthose related to memory loss [3, 4].Results and Conclusion: We found that both non-predis-posed and predisposed organoids exhibited active sponta-neous neuronal function and network activity. Exposure ofthese organoids to glycine induced a persistently enhancedrate of neuronal spikes, a physiological correlate ofmemoryformation called Long-Term Potentiation (LTP). Exposureof these organoids to glutamate/glycine induced LTP aswell as Long-Term Depression (LTD), a physiological cor-relate of memory loss. Overall, we observed differencesbetween the predisposed and non-predisposed organoidsin neuronal excitability and response to depressants which

may influence excitotoxicity events and level of fatigue inneuronal activity in these organoids.

References

[1] Lancaster, et al. Nat Protoc. 2014;9(10):2329–2340.[2] Renner, et al. Embo J. 2017;36(10):1316–1329.[3] Igartua, et al. Neuropharmacology. 2007;52(8):1586–

1595.[4] Hasegawa, et al. Sci Rep. 2015;5:7707.

82. The celecoxib derivative AR-12 inducesautophagy and controls prion infection in vitro andin vivo

Basant A. Abdulrahmana,b, Dalia Abdelaziza,c, SimrikaThapaa,c, Li Lua,c, Sabine Gilchb,c and Hermann M.Schatzla,c

aDepartment of Comparative Biology & Experimental Medicine,Faculty of Veterinary Medicine, University of Calgary, Calgary,Canada; bDepartment of Ecosystem & Public Health, Faculty ofVeterinary Medicine, University of Calgary, Calgary, Canada;cCalgary Prion Research Unit, and Hotchkiss Brain Institute,University of Calgary, Calgary, Canada

CONATCT Basant A. Abdulrahman [email protected]

ABSTRACT

Prion diseases are fatal infectious neurodegenerativedisorders that affect both humans and animals. Theautocatalytic conversion of the cellular prion protein(PrPC) into the pathologic isoform PrPSc is a keyfeature in prion pathogenesis. AR-12 is an IND-approved derivative of celecoxib that demonstratedpreclinical activity against several microbial diseases.Loss of neurons, astrogliosis and mild microglia activa-tion are the main pathological features of prion dis-eases. This results in a progressive spongiformdegeneration of the central nervous system (CNS),leading to ataxia, behavioural changes and, in humans,highly progressive loss of intellectual abilities. In thelast two decades, great efforts have been made to estab-lish treatment options for prion diseases. Theseincluded testing existing drugs for anti-prion activityin experimental models with only a few agents progres-sing to human studies of patients with prion diseases.Investigations to date have not resulted in a recognized/proven treatment for prion diseases. Recently, AR-12(a.k.a. OSU-03012), a potent inhibitor of PDK-1 (phos-phoinositide-dependent kinase 1) and autophagy sti-mulator, has been shown to facilitate clearance ofmisfolded proteins. The latter proposes AR-12 to be a

42 ABSTRACT

potential therapeutic agent for neurodegenerative dis-orders. In our study, we investigated the role of AR-12in controlling prion infection. We tested AR-12 inprion infected neuronal cells (ScN2a, ScCAD5) andnon-neuronal cells (ScMEF). Immunoblotting and con-focal microscopy results showed that AR-12 signifi-cantly reduced PrPSc levels after only 72 h oftreatment. Furthermore, infected cells were cured ofPrPSc after exposure of AR-12 for only 2 weeks. Wepartially attribute the influence of the AR-12 on prionpropagation to autophagy stimulation. Furthermore, wetested the therapeutic effect of AR-12 in vivo in RML-infected mice. Interestingly, treatment with AR-12(intraperitoneally and orally) significantly extendedthe survival of the treated animals compared to thecontrol group, in line with our previous findings thatdrug-induced stimulation of autophagy has anti-prioneffects in vitro. Taken together, this study demonstratesthat AR-12 represents a potential new therapeutic agentfor prion diseases and possibly protein misfolding dis-orders involving prion-like mechanisms.

KEYWORDS: Prions; PrPSc; Autophagy; AR-12

83. Rationally designed, structure-based vaccinecandidates targeting prion diseases

Andrew Fanga, Xinli Tanga, Brian Tancownya,Xiongyao Wanga* and Holger Willea

aCentre for Prions and Protein Folding Diseases & Department ofBiochemistry, University of Alberta, Edmonton, Alberta, Canada

CONTACT Andrew Fang [email protected]

*Present address: School of Materials Science andEngineering, Harbin Institute of Technology, Weihai,Shandong, China

ABSTRACT

Prion diseases are caused by the misfolding of PrPC toPrPSc, resulting in radical changes in secondary andtertiary structure. The lack of prophylactic or therapeu-tic vaccines means uncontrolled spread and an invari-ably fatal outcome for the host. No high-resolutionstructure for PrPSc exists despite it having been studiedextensively, but there is good evidence to suggest afour-rung β-solenoid structure. HET-s is a fungalprion protein that shares the β-solenoid fold withPrPSc, albeit with only two-rungs. A four-rung β- sole-noid version of HET-s, termed HET-2s, was engineeredvia a linker connecting the prion-forming domain ofHET-s twice. Strategic placement of seven PrP-based

amino acids into HET-2s allowed us to mimic the sur-face structure of PrPSc and marks a new, structure-based approach in prion vaccine design. Constructswere expressed in E. coli, purified, and assembled intoamyloid fibrils morphologically indistinguishable fromnative HET-s fibrils. Vaccine candidates showing goodfibrillization, and therefore adopting the proper β-sole-noid fold, were used for immunization experiments.We recently showed that one of our vaccine candidatesresulted in a specific immune response towards nativePrPSc.

Efficacy testing using an oral prion infectionmodel in Syrian hamsters is ongoing to evaluatewhether the vaccine candidate is effective againstperipheral infection routes. Monoclonal antibodies(mAbs) that originated from the immunized micewere shown to recognize native PrPSc in brainhomogenates from all prion isolates that weretested. A competition ELISA that uses these mAbsis showing great potential as a diagnostic tool (SeeTang et al. poster). Moreover, the ability of thesemAbs to recognize native PrPSc from various strainsalso reveals shared structural characteristics (SeeWille et al. poster). Furthermore, the IgM and IgGclones are being sequenced and revealed uniquecomplementarity-determining regions, indicatingthey all have different epitopes (See Rathod et al.poster). Lastly, this approach has been used to alsodesign vaccines against other amyloid diseases, spe-cifically targeting misfolded Aβ, α-synuclein and tauprotein (See Flores-Fernandez et al. poster). Takentogether, our novel vaccine design moves us onestep closer to develop passive and active immu-notherapies for the prion diseases.

84. Stress granules “road to ruin’’ inneurodegenerative diseases

Neelam Younas, Saima Zafar and Inga Zerr

Department of Neurology, Clinical Dementia Centre, UniversityMedical Centre Goettingen(UMG), Germany

ABSTRACT

Introduction: Stress granules are membrane less cyto-plasmic foci of mRNA and RNA binding proteins thatare formed in the cell to cope with variety of stressors.Emerging evidences have shown disturbances in thedynamics of these non-membranous foci, as a patho-physiological element of amyotrophic lateral sclerosis(ALS), frontotemporal dementia (FTD), and more

PRION 43

recently in Alzheimer disease1,2. These evidences redir-ect research efforts towards understanding the role ofstress granule biology in neurodegenerative diseases. Inthis study, we aim to explore the role of oxidative-stressto proteopathic disease linked proteins which are majorhallmarks of neurodegenerative diseases.Methods: For generation of Cell Stress Model, humancervical adenocarcinoma (HeLa) cell line was treated with0.6mM sodium arsenite (a classical stress inducer) at 37°Cfor 60min. Immunofluorescence was used for visualizationof stress granules. Differential expression of selected targetproteins was analysed in untreated (control) and treatedcells (stressed) and in Human brain patients byImmunoblotting. Tau-Pathology Model was used to findout direct mechanistic link of target proteins to diseasepathology.Results: Interestingly, we found that aggregated proneproteins particularly phosphorylated form of Tau andPrion protein (Tau, PrP) were recruited to cytoplasmicstress granules after sodium arsenite treatment. Stressgranules were identified by classical marker TIA-1 andPABP. To find out stress specific interactors of aggre-gated prone proteins interactome mapping of theseproteins in untreated and stress induced cells revealeda common stress specific target, splicing factor proline,and glutamine rich (SFPQ) which is an importantcomponent of Neuronal transport granules. Here, weshow that SFPQ is also a constituent of cytoplasmicstress granules. Arsenite treatment induced cytoplasmicSFPQ granules. Further to find out granule formationpropensity of SFPQ, it was subjected to catGRANULEsoftware which predicted a score of 1.66 showing agood propensity of SFPQ to form granules or liquid-liquid demixing. SFPQ was found to be significantlyregulated in Alzheimer and CJD patient’s brain in sub-type dependent manner.Conclusions: Our results highlight novel association/localization of aggregation prone proteins with stressgranules linking cellular stress response to pathobiologyof these proteins. Further differential regulation ofSFPQ, its LLPS property and its localization in stressgranules and interaction with Tau and Prion Proteinlinks dots for SFPQ as might be crucible of aggregationvia stress granules.

Supported by Helmholtz-Alberta Initiative –Neurodegenerative Diseases Research (HAI-NDR);Alberta innovates Bio solutions, and DZNE Goettingen.

References

[1] Zhang P, et al. 2018;bioRxiv: 348,870.[2] Vanderweyde, et al. Cell Reports. 2016;15:1455–1466.

85. Kinetics of prion seeding activity in brains ofmice infected with mouse-adopted BSE prion

Yoshifumi Iwamaru, Yuichi Matsuura and KohtaroMiyazawa

Prion Disease unit, Division of Transboundary Animal Disease,National Institute of Animal Health (NIAH), National Agricultureand Food Research Organization (NARO), Tsukuba, Japan

CONTACT [email protected]

ABSTRACT

Prion diseases are fatal neurodegenerative disorders inhumans and animals. The key event in the pathogenesisof these diseases is the conversion of host-encodednormal cellular prion protein (PrPC) into its patho-genic isoform (PrPSc) and its accumulation in thecentral nervous system. PrPSc is copurified with infec-tivity and thus thought to be the main constitute of theagent of prion diseases. PrPSc is partially resistanttowards the proteinase K (PK) digestion and PK-resis-tant PrPSc (PrPres) has been used extensively as asurrogate marker for prion infection. However, severalstudies have revealed that prion infectivity is not alwayswell-correlated with total PrPres levels. Real-time quak-ing-induced conversion (RT-QuIC) is one of newlydeveloped in vitro amplification methods based onprion-seeded amyloid fibril formation by recombinantprion protein. According to a previous study, prionseeding activity appears to be correlated with prioninfectivity than with total PrPres-levels. In our previousstudy of kinetics of mouse-adopted BSE prion propaga-tion and PrPres accumulation, we demonstrated thatthe brain titer reached a plateau at 100 days post infec-tion whereas PrPres continuously increased towards theend of the incubation period. In this study, we haveproceeded to measure kinetics of prion seeding activityin the brain of these mice. We have used end-pointdilution RT-QuIC on brain homogenates to produce adetailed measurement of seeding activity throughoutthe incubation period of the mice and evaluated thetemporal relationships among seeding activity, infectiv-ity, and PrPres levels.

86. Characterization of cellulose ether liposomesfor the inhibition of prion formation in prion-infected cells

Keiko Nishizawaa, Kenta Teruyaa, Ayumi Ogumaa, YujiSakasegawaa, Hiroshi Kamitakaharab, HermannSchatzlc, Sabine Gilchd and Katsumi Doh-uraa

aDepartment of Neurochemistry, Tohoku University Graduate School ofMedicine, Sendai, Japan; bDivision of Forest & Biomaterials ScienceGraduate School of Agriculture, Kyoto University, Kyoto, Japan;cDepartment of Comparative Biology and Experimental Medicine,

44 ABSTRACT

Faculty of Veterinary Medicine; Hotchkiss Brain Institute; University ofCalgary, Canada; dDepartment of Ecosystem and Public Health, CalgaryPrion Research Unit, Faculty of Veterinary Medicine, Hotchkiss BrainInstitute, University of Calgary, Calgary, Canada

CONTACT Katsumi Doh-ura [email protected]

ABSTRACT

Our previous study demonstrated that a single per-ipheral administration of cellulose ethers (CE) dose-dependently prolonged the survival of prion-infected rodents when immediately administeredafter prion infection or even 1 year prior to infec-tion. Additionally, CE inhibited prion formation ofpersistently prion-infected cells in a manner parallelto the in vivo efficacy. However, the mechanism ofanti-prion action of CE is not fully understood.Even in the most effective CE, considerable doses,such as few mg/gm body weight in rodents or tensof mg/ml in cell culture media are needed forobtaining substantial effects. Therefore, improve-ment of the bioavailability is required for the prac-tical use of CE. In this study, we prepared CE-loaded liposomes and examined the anti-prion activ-ity in prion-infected cells to compare the effect ofCE-loaded liposomes with that of unformulated CEon anti-prion activity. We also modified CE-loadedliposomes and investigated the uptake levels inprion-infected and macrophage-like cells, and thencompared the uptake levels with anti-prion activitylevels to help elucidate the anti-prion mechanism ofCE-loaded liposomes. The liposomal formulationreduced the EC50 dose of CE by <1/200-fold inprion-infected cells. Compared to empty liposomes,CE-loaded liposomes were taken up much morehighly by prion-infected cells and less by macro-phage-like cells. Phosphatidylserine modificationreduced the uptake of CE-loaded liposomes inprion-infected cells and did not change the anti-prion activity, whereas increased the uptake inmacrophage-like cells. Polyethylene glycol modifica-tion reduced the uptake of CE-loaded liposomes inboth types of cells and reduced the anti-prion activ-ity in prion-infected cells. These results suggest thata liposomal formulation of CE is more practicalthan unformulated CE and showed that the CE-loaded liposome uptake levels in prion-infectedcells were not associated with anti-prion activity.Although further improvement of the stealth func-tion against phagocytic cells is needed, the liposomalformulation is useful to improve CE efficacy andelucidate the mechanism of CE action.

87. Nascent β structure in the elongatedhydrophobic region of a Gerstmann-Sträussler-Scheinker PrP Allele

Ze-Lin Fua,b, Peter C. Holmesb, David Westawaya,b andBrian D. Sykesb

aCentre for Prions and Protein Folding Diseases, University ofAlberta, Edmonton, AB Canada; bDepartment of Biochemistry,University of Alberta, Edmonton, AB Canada

ABSTRACT

Objective: To understand the molecular basis of themisfolding of a novel prion protein (PrP) mutant thatcauses Gerstmann-Straussler-Scheinker (GSS) disease.This mutant designated ‘HRdup’ was defined in a 34-year-old GSS patient and contains an 8-amino-acidduplication (LGGLGGYV) after Val129. This alleleencompasses both part of the glycine-rich portion ofthe hydrophobic region (HR) and all the first beta strandand is thus distinct from other GSS mutations thatcomprise missense mutations or insertions of extraoctarepeats. Transgenic (Tg) mice expressing HRdupPrP develop a spontaneous neurological syndrome andproteinase K digestion of brain homogenates from thesemice shows a pathognomonic GSS band at ~8kDa [1].Methods: For structural analysis, we purified 13C, 15Ndouble-labelled recombinant mPrP HRdup (118–231)along with two control alleles, mPrP V128 (118–231)and mPrP WT (118–231). Using 3D NMR experimentsperformed at pH 5.0 and in the absence of added co-factors, we assigned the NMR spectra of both HRdupand V128; therefore, determined the secondary struc-ture of these two proteins and explored the molecularmotions and dynamics of HRdup and V128. The sec-ondary structure prediction programmes and MDsimulation were also employed to predict and verifythe conformation of the N-terminus of HRdup.Results: The NMR experiments showed that HRduphas the same canonical globular PrPC structure asV128, and that the elongated N-terminus of HRdupdisplayed significant β-propensity while maintainingits high flexibility. MD simulation depicted this nascentβ-structure in the hydrophobic region as a β-turn. Inaddition, we observed that a methionine/valine poly-morphism at codon 128 (equivalent to codon 129 inhumans) in the presence of oligomerization facilitatedby high protein concentrations affected conformationalexchange dynamics at residue G130 [2].Conclusions: We conclude that the novel β-structure atthe elongated hydrophobic region of HRdup is the rootcause of spontaneous misfolding of HRdup PrP. As theexchange dynamics of G130 also suggests the potential

PRION 45

involvement of the canonical β-sheet in the initializa-tion of PrP misfolding, we hypothesize that the hydro-phobic region of HRdup can adopt a fully extendedconfiguration, fold back and develop into a three-strand antiparallel β-structure that incorporates thecanonical β-sheet. This proposed structure is highlyconsistent with the structure observed in full lengthhPrP when crystalized with nano-body at its N-term-inal end [3].

References

[1] Mercer RCC, et al. PLOS Pathogen. 2018;14(1):e1006826.

[2] Fu Z-F, et al. Submitted for publication.[3] Abskharon RNN, et al. JACS 2014;136(2):937–944.

88. Sporadic CJD in pregnancy: an interdisciplinarysuccess story

Collin Luka, Allison Thieleb, Christina Orru-Grovemanc,Byron Caugheyc and Valerie L. Sima,d

aDepartment of Medicine, Division of Neurology, Faculty ofMedicine & Dentistry, University of Alberta; bDepartment ofObstetrics and Gynaecology, Division of Maternal Fetal Medicine,Faculty of Medicine & Dentistry, University of Alberta; cRockyMountain Laboratories, NIAID, National Institutes of Health,Hamilton, MT, USA; dCentre for Prions and Protein FoldingDiseases, University of Alberta

CONTACT Valerie L. Sim [email protected]

ABSTRACT

Sporadic Creutzfeldt-Jakob disease (sCJD) affects 1–2people per million per year, with a peak incidence inthe 7th decade of life. Rarely, CJD can affect women ofchildbearing age.

There are seven previously reported cases of CJD (sixsporadic, one iatrogenic) in pregnancy with five healthydeliveries and no evidence of vertical transmission todate.1–5 In these reports, immunohistochemical screensof placental tissue were negative for PrPSc, but in the onestudy that tested for placental infectivity, prion diseasewas transmitted to five of eight cerebrally inoculated Balbmice.2 The possibility that placenta may contain infec-tious prions has implications for vertical transmission andrisk management of delivery. We present a 35 year oldG2P1 woman with probable sCJD who presented in her10th gestational week with a 7-month history of progres-sive memory impairment and visuospatial difficulties.Initial investigations, including CSF studies, were negativefor autoimmune, inflammatory, infectious, and neoplasticconditions. A positive ds-DNA raised the possibility of

lupus, for which she was started on Plaquenil, but noother clinical criteria were identified. An electroencepha-logram demonstrated bilateral diffuse slowing. BrainMRIwas delayed until the beginning of the 2nd trimester outof theoretical concern for foetal exposure. The MRIshowed restricted diffusion of basal ganglia and corticalribboning consistent with Creutzfeldt-Jakob disease. Arepeat CSF revealed elevated 14–3-3 (>80 000 AU/mL),elevated Tau protein (>10 513 pg/mL) and positive EP-QuIC assay. There was no PRNP gene mutation. The rateof clinical progression required extensive interdisciplinarymanagement to preserve the pregnancy and safely planfor a number of possible obstetric scenarios. A Kaofeedwas required to maintain nutrition. Oxygen was providedby nasal prongs; intubation and ventilation was notrequired. Fevers and tachycardia developed but withoutevidence of infection, suggesting central autonomic dys-regulation. Ultimately, a healthy baby was delivered via C-section at 36 weeks 3 days gestation, with no obstetric orpost-anaesthetic complications. Cord blood, amnioticfluid and placental tissue were collected for evaluationby RT-QuIC. The patient passed away 24 days after deliv-ery. A discussion of diagnostic challenges, managementstrategies and QuIC results will be presented.

References

[1] Xiao X, et al. Am J Pathol. 2009;174(5):1602–1608[2] Tamai Y, et al. N Engl J Med. 1992;327(9):649.[3] Sperling R, et al. J Obstet Gynecol Neonatal Nurs.

2005;34(5):546–550.[4] Di Gangi S, et al. J Matern Fetal Neonatal Med. 2015;28

(3):254–261.[5] Dalmas AF, et al. Ann Fr Anesth Reanim. 2010;29

(11):815–817.

89. Exploiting their sweet tooth: sugars againstprions

M. Kumara,b, Garrett J.B. Molnerc and Valerie L. Sima,b,aDepartment of Medicine, Division of Neurology, Faculty ofMedicine & Dentistry, University of Alberta; bCentre for Prions andProtein Folding Diseases, University of Alberta; cDepartment ofMedical Microbiology, Faculty of Medicine & Dentistry, Universityof Alberta

CONTACT M. Kumar [email protected]

ABSTRACT

Endoplasmic reticulum-associated degradation(ERAD) is critical to cellular protein homoestasis.The accumulation of unfolded or misfolded proteinsin the ER challenges this proteostasis and has been

46 ABSTRACT

implicated in neurodegeneration1. In prion disease,cellular prion protein (PrPC) is converted into a β-sheet rich infectious form (PrPSc)2. PrPC is a mem-branous protein that is variably N-glycosylated and,like other glycoproteins, depends on enzymes such asglucosidase I, II and glucosyltransferase for matura-tion of sugar residues as well as proper folding of theprotein and its ultimate localization. Failure to prop-erly glycosylate a membrane protein within the ERinduces proteostasis stress in the ER and activates ERquality control machinery1,3,4. We hypothesize thattrimming the glycans during PrPC synthesis willincrease PrP levels in the ER and activate ER-asso-ciated degradation (ERAD). In the context of prioninfection, this could both reduce available substratefor conversion and enhance clearance of misfoldedPrP. We inhibited α- and β-glucosidases with foursugar analogues in CAD5 cells chronically infectedwith RML strain prions. Moderate dosages of indivi-dual analogues resulted in complete eradication ofPrPSc within 72 h. Interestingly, treating with lowerdosages in combination generated different PK-resis-tant prion species. Immunoblotting experiments alsoindicated a change in the protein level of OS-9, amajor player in ER quality control and ERAD. OS-9binds terminally misfolded non-glycoproteins as wellas improperly folded glycoproteins and transfersthem to ubiquitination machinery for targeted degra-dation5-7. We speculate that truncation of N-linkedglycans on PrP with the combination of lower dosesugar analogues resulted in the formation of modifiedimproperly folded mono- and di-glycosylated PrPCwith altered conformational repertoire. These typesof sugar analogues have high oral bioavailability, caneasily penetrate the central nervous system, haveextensively characterized biological and pharmacoki-netic properties. As such, they may have therapeuticutility for prion diseases at moderate dose. In addi-tion, they represent a novel tool for the experimentalmanipulation of prion strains.

References

[1] Rendleman, et al. eLife. 2018;7:e39054[2] Prusiner SB. Proc Natl Acad Sci. 1998;95:13,363–13,383[3] Fagioli, et al. J Biol Chem. 2001;276:12,885–12,892;4.[4] Smith, et al. Science. 2011;334:1086–1090.[5] Bernasconi, et al. J Biol Chem. 2008;283:16,446–

16,454;6.[6] Hosokawa, et al. J Biol Chem. 2009;284:17,061–17068.[7] Alcock, et al. J Mol Biol. 2009;385:1032–1042.

90. Estimation of the binding sites of inhibitoryfactors on recombinant PrP molecule by RT-QuIC

Akio Suzuki, Kazuhei Sawada, Takeshi Yamasaki andMotohiro Horiuchi

Laboratory of Veterinary Hygiene, Faculty of Veterinary Medicine,Hokkaido University, Sapporo, Japan

CONTACT Akio [email protected]

ABSTRACT

Background: Real-time quaking-induced conversion(RT-QuIC) is one of several highly sensitive methodsfor the detection of prions. However, the reaction iseasily interfered by inhibitory factors in tissue homo-genates. We previously showed that recombinant cervidPrP (rCerPrP) can react with prions even in the pre-sence of tissue homogenates (Prion 2017, P152). Toclarify the region of the PrP molecule that is affectedby inhibitory factors, we produced a variety of rPrPmutants and analysed the reactivity of the PrP mutantsto H- and L-BSE prions by RT-QuIC.Material and Methods: Full-length recombinant sheep(ARQ) PrP (rShPrP), mouse PrP (rMoPrP), rCerPrP, andtheir C-terminal chimeras (rCerPrP-MoCterm andrMoPrP-CerCterm), and their mutants that have point-mutations at ShPrP-specific amino acid (aa) 98 S and/orCerPrP-specific 173 N and 177 T (rMoPrP-S169NCer/N173TCer, rMoPrP-T94SSh/N96SSh/S169NCer/N173TCer

and rCerPrP-N173SMo/T177NMo/, rCerPrP-T98SSh/N173SMo/T177NMo) were used as substrates. Brainhomogenates (BH) of cattle infected with H-BSE and L-BSE were serially diluted with PBS or normal cattle BHand used as seeds. RT-QuIC reactions were performedunder conditions containing 10 mM Na-phosphate (pH7.4), 500 mM NaCl and 0.001% SDS at 37 °C.Results: rShPrP, rMoPrP and rCerPrP could comparablydetect H-BSE and L-BSE diluted at 10−4–10−7 with PBS.However, the reaction of rShPrP with L-BSE was delayedfor 13.4 h on average and those of rMoPrP with H-BSEand L-BSE were delayed for 27.4 h and completely inhib-ited by 0.1% normal BH, respectively. Compared toauthentic rPrPs, the reaction of rCerPrP-MoCterm withL-BSE in the presence of normal BH was delayed for10.7 h, whereas that of rMoPrP-CerCterm was shortenedby 21.9 h. These results suggest that the C-terminus ofrMoPrP is more affected by the inhibitory factors com-pared with those of rCerPrP. On the other hand, thereaction of rCerPrP-N173SMo/T177NMo/and rCerPrP-T98SSh/N173SMo/T177NMo with L-BSE was delayed for10.8 h and 13.8 h compared to rCerPrP, whereas those ofrMoPrP-S169NCer/N173TCer and rMoPrP-T94SSh/

PRION 47

N96SSh/S169NCer/N173TCer was shortened by 11.5 h and14.0 h compared to rMoPrP. Taken together, the inhibi-tory factors may also interfere with the region includingCerPrP specific N173, and T177.Conclusion: Our results suggest that conversion effi-ciency in the RT-QuIC reaction may be interfered withby the inhibitory factors at least at two regions of thePrP molecule, one is the C-terminus and the other isthe region including CerPrP-specific aa 173 N and177 T. These regions have been showen to be adjacentin the three-dimensional structure of PrPC; therefore,inhibitory factors may bind to the site including theseregions.

91. PrP regulates neural stem cell senescencethrough epidermal growth factor receptor

Bradley R. Groveman, Simote T. Foliaki, Aleksandar R.Wood, Brent Race and Cathryn L. HaighRocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, USA

CONTACT Bradley R. Groveman [email protected]

ABSTRACT

The prion protein (PrP) is expressed in a wide varietyof progenitor cells and is known to be involved inneural stem cell self-renewal processes. The mechan-isms by which PrP acts to modulate cell homeostasishave, however, not been fully elucidated. Recent stu-dies have shown that PrP has a direct role in prevent-ing premature senescence [1], and that knockdown ofPrP increases senescence markers and decreases thecell proliferative potential. PrP has also been shown toregulate epidermal growth factor receptor (EGFR)through Notch signalling. Notch signalling, whichcan upregulate EGFR, is disrupted in the absence ofPrP [2], leading to a reduction in EGFR expression.Furthermore, inhibition of EGFR or depletion of EGFhave been shown to suppress senescence in prolifera-tive cells [3]. Herein we attempt to show the role ofPrP in regulating cellular senescence through theEGFR pathway.

KEYWORDS: Prion; PrP; neural stem cell; senescence;epidermal growth factor receptor; EGFR

References

[1] Boilan E, et al. Mech Ageing Dev. 2018;170:106–113.[2] Martin-Lannerée S, et al. Stem Cells. 2017;35(3):754–

765.[3] Alexander PB, et al. Cell Res. 2015;25(1):135–138.

92. Region-specific interference of PrPSc

accumulation in spleen and brain by poly-L-arginine (PLR)

Jieun Kima, Taeyeon Kimb, A-ran Kima, Hakmin Leea,Trang H. T. Trinha, Junwu Shina, Sungeun Leea andChongsuk Ryoua

aDepartment of Pharmacy, College of Pharmacy and Institute ofPharmaceutical Science and Technology, Hanyang University,Ansan, Gyeonggi-do, Republic of Korea; bDivision ofDevelopmental Biology and Physiology, School of Biosciences andChemistry, Institute for Basic science, Sungshin University, Seoul,Republic of Korea.

CONTACT Chongsuk Ryou [email protected]

ABSTRACT

Background: In our previous study, PLR was shown toinhibit prion propagation in the cell culture model ofprion diseases (1). In current study, we investigated theeffect of PLR in vivo whether it inhibits prions.Material and Methods: Firstly, mice inoculated intra-peritoneally (IP) with prions were administered withPLR twice a week for 4 weeks 10–40 mg/kg by the IProute. Spleen were collected from infected mice at every21-day until 84 days post-inoculation. In anotherapproach, to observe the delay of disease onset, miceinfected intracerebrally with prions, were administeredwith PLR twice a week for 4 weeks 10–25 mg/kg PLR bythe IP route. As disease onsets, the brain and spleen werecollected. The level of PrPSc in the brain and spleen weredetected by Western blotting. The accumulation of thePrPSc and glial fibrillary acidic protein were monitoredusing immunohistochemistry. Vacuolation in the brainwas investigated using H&E stained brain sections.Result: PrPSc propagation was suppressed in the spleen ofPLR-treated group during the asymptomatic stage.Administration with 10–25 mg/kg PLR prolonged theincubation time of prion diseases over seven days. Atthe disease onset, the level of PrPSc reduced in the spleen,but unchanged in the brain. However, immunohisto-chemistry analysis showed that the level of PrPSc inwhite pulp of the spleen and specific areas of the brain,such as hypothalamus, cortex, and hippocampus, but notmid-brain, thalamus, and straitum, was reduced by PLR.Moreover, glial fibrillary acidic protein staining indicatedthat PLR decreased astrogliosis in thalamus, cortex, andpolymorph layer 3 of cerebral cortex of brain but not inother brain regions. In addition, H & E staining indicatedthat the decrease of vacuolation area and vacuole countsin mid-brain, hypothalamus, hippocampus, and straitumregions of the brains from the PLR-administered mice.Conclusion: Considering suppression of prion propa-gation in spleen during asymptomatic stage, significant

48 ABSTRACT

increase of survival time and brain region-specific inhi-bition of neuropathological onsets, we demonstratedPLR as a potential therapeutic candidate for prion dis-eases and the value of further studies.

KEYWORDS: Prion; poly-L-arginine; PrPSc; spleen;brain

Reference

[1] Waqas, et al. Mol Cell Biochem. 2017;428(1–2):57–66.

93. Prion protein distribution among white-taileddeer and mule deer exocrine glands

Anthony M. Nessa,b, Kelsey Saborakic, Camilo D.Velásqueza,b, Danielle Gushuea,b, Alicia O. Garciaa,b,Joshua Craiga,b, Taraneh Ashrafia,d, Judd Aikena,d,Susan Linglec*, Debbie McKenzie1,2*

aCentre for Prions and Protein Folding Diseases, University of Alberta;bDepartment of Biological Sciences, University of Alberta;cDepartment of Biology, University of Winnipeg; dDepartment ofAgricultural, Food, and Nutritional Sciences, University of AlbertaCONTACT Anthony M. Ness [email protected]

ABSTRACT

Background: Chronic wasting disease (CWD) is a conta-gious neurodegenerative prion disease afflicting cervids inCanada, the United States, South Korea, and Scandinavia.The precise mechanism of natural CWD transmission ispoorly understood but is believed to involve both directcontact between cervids and exposure to environmentalreservoirs of PrPCWD. The scent glands in the face andlegs of deer are numerous and serve for direct and indirectinter-individual communication and social behaviours.Glandular innervation by the autonomic nervous systemmay act as a site forCWDprion uptake, neuroinvasion, andretrograde transport towards the central nervous system.Materials and Methods: Facial and leg exocrine glandswere collected from adult and white-tailed deer andmule deer males and females harvested fromCanadian Forces Base Wainright, Alberta, Canada.Vomeronasal, forehead, pre-orbital, parotid, and nasalglands in the head, and the tarsal, metatarsal, andinterdigital glands in the legs were homogenized forbiochemical analysis or embedded for histological ana-lysis. White-tailed deer and mule deer glandular PrPC

levels were determined by Western blot and ELISA.The distribution of PrPD was evaluated by immunohis-tochemistry (IHC).Results and Discussion: We show the relative PrPC

levels and distribution in the various facial and legexocrine glands studied. IHC revealed PrPD withinselect glands. The involvement of cervid exocrine

glands in CWD transmission has been poorly studied;however, our observations suggest these glands act assites for prion entry and shedding – deserving furtherinvestigation. Cervid exocrine glands may represent abiologically and ecologically important transmissionroute of CWD in deer. Further, concurrent researchsuggests that species-and sex-specific social behaviours,involving the glandular tissues, may provide insights asto why CWD is most prevalent in mule deer males andleast prevalent in white-tailed females.

KEYWORDS: Chronic wasting disease; prion; deer;cervids; glands; transmission

94. Plasma tau and neurofilament light chain asbiomarkers for the human prion diseases:evaluation in a large prospective natural historycohort

Andrew G. B. Thompsona, Prodromos Anastasiadisa,Akin Nihata,b, Tze Mok Howa,b, Peter Rudgea,b,Amanda Heslegravec, Henrik Zetterbergc, JohnCollingea,b, Graham S. Jacksona and Simon H. Meada,b

aMRC Prion Unit, University College London, UK; bNational PrionClinic, University College London Hospitals NHS Foundation Trust,UK; cDementia Research Institute Biomarkers Laboratory, UniversityCollege London, UK.

CONTACT Andrew GB Thompson

ABSTRACT

Background: Diagnostic methods for prion diseasehave advanced substantially, but unmet needs remainfor biomarkers to fill a number of other importantroles. These include monitoring of disease activity, pre-dicting symptom onset in individuals at high risk (suchas carriers of prion protein gene (PRNP) mutations),and enabling earlier diagnosis. Tau and neurofilamentlight chain (NfL) protein concentrations in blood areknown to be elevated in prion disease, and have shownpromise as quantitative markers of central nervoussystem disease activity in a range of other conditions.Materials and Methods: We used ultrasensitiveimmuno-assays (Simoa) to measure tau and NfL con-centrations in 709 plasma samples taken from 377 indi-viduals with prion disease during their participation in alarge prospective clinical study (the UK National PrionMonitoring Cohort), in addition to healthy and neuro-logical control groups. This provides an unprecedentedopportunity to analyse their clinical associations andpotential as biomarkers.Results: Plasma tau and NfL concentrations areincreased across all prion disease types. For distinguish-ing sporadic CJD (sCJD) from a clinically relevant

PRION 49

‘prion mimic’ control group, both show moderate diag-nostic value (area under ROC curves: tau 0.81, NfL0.72). In sCJD, plasma NfL was substantially elevatedin every sample tested, including during early diseasewith minimal functional impairment. Plasma tau wasindependently associated with rate of clinical progres-sion in sCJD, while plasma NfL showed independentassociation with severity of functional impairment. Inasymptomatic PRNP mutation carriers, plasma NfL washigher in samples taken within 2 years of symptomonset than in those taken earlier. We present biomarkertrajectories for 9 PRNP mutation carriers who devel-oped symptoms during follow-up, and show potentialfor plasma NfL as a ‘proximity marker’.Conclusions: We conclude that plasma tau and NfLrepresent the best currently available candidates for anumber of vital biomarker roles in prion disease. Wediscuss directions for future work, particularly focuss-ing on ways that these biomarkers could be valuable fortherapeutic research.

95. Gene-edited murine cell lines for propagationof chronic wasting disease prions

Rupali Waliaa,b, Rohit Chandraa,c and Hermann M.Schatzla,b

aDepartment of Comparative Biology & Experimental Medicine,University of Calgary, Calgary, Canada; bCalgary Prion ResearchUnit, University of Calgary, Calgary, Canada; cFaculty of Science,University of British Columbia, Vancouver

CONTACT Rohit Chandra [email protected]

ABSTRACT

Chronic wasting disease (CWD), a highly contagious priondisease, has recently emerged as a threat to both farmedand free ranging cervids across North America, SouthKorea, and Europe. There is thus a pressing need to designversatile cell culture models which can complement theanimal studies on CWD and help to contain this disease.Defining such cell culture models is mostly a randomprocess, and includes extensive subcloning and selection.To extend the range of cell models propagating CWDprions, we gene-edited mouse cell lines known to wellpropagate murine prions. Endogenous prion protein(PrP) was ablated in CAD5 cells and immortalized mouseembryonic fibroblasts (MEF), using CRISPR-Cas9 geneediting. PrP knock-out cells were reconstituted withmouse, bank vole (BV) and cervid PrP genes by lentiviraltransduction. Reconstituted cells expressing mouse PrPprovided proof-of-concept for re-established prion infec-tion. The promiscuous nature of BV PrPc was exploited todemonstrate the permissiveness of these reconstituted cell

lines to infection by CWD prions, even without subclon-ing. Cells reconstituted with BV or cervid PrP and infectedwith two types of CWD prions, white-tailed deer and muledeer prions, tested positive in prion conversion assay,whereas non-reconstituted cells were negative. We hereprovide a successful strategy for designing cell lines withspecific as well as broader susceptibility to prion infection,while retaining a common host background. Data obtainedby infecting such gene-targeted and re-constituted culturedcells will provide important new insights into species andprion strain barriers and provide new experimental sys-tems for studying CWD prions in vitro.

96. PrPSc aggregation state dictates biochemicalproperties of prions and disease pathogenesis

Sheng Chun Changa, Samia Hannaouia, StefanieCzubb and Sabine Gilcha

aDepartment of Ecosystem and Public Health, Faculty of VeterinaryMedicine, Calgary Prion Research Unit, Hotchkiss Brain Institute,University of Calgary, Calgary, Canada; bCanadian Food InspectionAgency Lethbridge Laboratories, Lethbridge, Canada

CONTACT Sheng Chun Chang [email protected]

ABSTRACT

Background: Transmissible spongiform encephalopa-thies (TSE) are devastating neurodegenerative diseasescaused by the conversion of the host-encoded cellularprion protein (PrPC) into the pathological misfolded iso-form known as PrPSc. Various strains of prions exist, eachof which consists of a unique conformational, biochem-ical, and biological profile. Previous studies have indicatedthat the profile of PrPSc aggregation states also is strainspecific. Our study investigates whether biochemical andbiological properties of individual fractions of PrPSc arestable upon serial passage in vivo.Methods: A pool of CWD2 elk brain homogenate wassubject to a sedimentation velocity gradient and separatedinto 30 fractions. Alternating fractions were intracerebrallyinoculated into transgenic mice overexpressing elk PrPC

(tgElk) and incubation times and clinical signs were mon-itored. Brains (first passage) were harvested upon the pro-gression of terminal prion disease. Brains of three miceeach inoculated with PrPSc fractions 2–6 (low molecularweight (MW)), 8–18 (medium MW), and 20–30 (highMW), respectively, were used for second passage intotgElk mice. Brain samples were subjected to digestionwith increasing concentrations of Proteinase K (PK) todetermine the PK concentration required to degrade 50%of PrPSc (cPK50).Results: In the first passage, mice inoculated with lowMW PrPSc aggregates exhibited symptoms of hyper-

50 ABSTRACT

excitability, while those inoculated with high MWaggregates were lethargic and gained weight, and aprolonged incubation was observed in both groups,while those given medium MW aggregates had adecreased incubation time. Biochemical analysisshowed that mice inoculated with low or mediumMW aggregates had PrPSc that were markedly lessresistant to PK digestion compared to the PrPSc frommice inoculated with high MW aggregates. These char-acteristics were no longer observed in the second pas-sage, where the incubation period and the PK resistanceamong the groups were similar, though the observedsymptoms in the mice inoculated with the low or highMW PrPSc were still hyper-excitability and lethargy,respectively, as in the first passage.Conclusion: Our study demonstrates that inoculationof different PrPSc aggregation states separated by mole-cular weight can lead to differing pathogenesis andtransient changes to the biochemical characteristics.This will add novel knowledge to our understandingof transmissible biochemical characteristics of prions.

97. In vitro seeding activity of glycoform-deficientprions from variably protease-sensitiveprionopathy and familial CJD associated withPrPV180I mutation

Zerui Wanga,b, Jue Yuana, Pingping Shena,b, RomanyAbskharonc, Yue Langa,b, Johnny Danga, AliseAdornatoa, Ling Xua, Jiafeng Chenb, Jiachun Fengb,Mohammed Moudjoud, Tetsuyuki Kitamotoe, JanLangeveldf, Brian Applebya,g,h, Jiyan Mac, QingzhongKonga,g,h, Robert B. Petersena,i, Li Cuib and Wen-QuanZoua,b,g,h

aDepartment of Pathology, Case Western Reserve University School ofMedicine, Cleveland, Ohio, USA; bDepartment of Neurology, The FirstHospital of Jilin University, Changchun, Jilin Province, the People’sRepublic of China; cCenter for Neurodegenerative Science, Van AndelResearch Institute, Grand Rapids, MI, USA; dNRA, Université Paris-Saclay,UR892, Virologie Immunologie Moléculaires, Jouy-en-Josas, France;eCenter for Prion Diseases, Tohoku University Graduate School ofMedicine, Sendai, Japan; fWageningen BioVeterinary Research,Lelystad, the Netherlands; gNational Prion Disease PathologySurveillance Center, Case Western Reserve University School ofMedicine, Cleveland, OH, USA; 8Department of Neurology, CaseWestern Reserve University School of Medicine, Cleveland, OH, USA;iFoundation Sciences, Central Michigan University College of Medicine,Mount Pleasant, MI, USA

CONTACT Wen-Quan Zou WQZ [email protected]

ABSTRACT

Both sporadic variably protease-sensitive prionopa-thy (VPSPr) and familial Creutzfeldt-Jakob diseaselinked to the prion protein (PrP) V180I mutation

(fCJDV180I) have been found to share a uniquepathological prion protein (PrPSc) pattern thatlacks the protease-resistant PrPSc glycosylated atresidue 181, apparently because two of four cellularPrP (PrPC) glycoforms are not converted into PrPSc.To investigate the seeding activity of these uniquePrPSc molecules, we conducted in vitro prion con-version experiments using serial protein misfoldingcyclic amplification (sPMCA) and real-time quak-ing-induced conversion (RT-QuIC) assays with dif-ferent PrPC substrates. Unexpectedly, we observedthat the seeding of PrPSc from VPSPr or fCJDV180I

in the sPMCA reaction with brain homogenatesfrom normal human or humanized transgenic (Tg)mice generated PrPSc molecules that are dominatedby the diglycosylated isoform, along with PrPSc

monoglycosylated at residue 181. The efficiency ofPrPSc amplification was significantly higher in MMthan in VV human brain homogenate, whereas itwas higher in TgVV than in TgMM mouse brainhomogenate. PrPC from the brain homogenate mix-ture of TgMM and Tg mice expressing PrPV180I

mutation (Tg180), but not that from TgV180Ialone, was converted into PrPSc by seeding withthe VPSPr or fCJDV180I. The RT-QuIC seedingactivity of PrPSc from VPSPr and fCJDV180I wassignificantly lower than that of sCJD. Our resultssuggest that the formation of glycoform-selectiveprions may be associated with an unidentified factorin the affected brain and the glycoform-deficiency ofPrPSc does not affect the glycoforms of in vitronewly-amplified PrPSc

Funding

Supported in part by the CJD Foundation and theNational Institutes of Health (NIH) NS062787 andNS087588 to W.Q.Z., NS062787 and NS109532 to W.Q.Z., and Q.K., NS088604 to Q.K., the Centers forDisease Control and Prevention Contract UR8/CCU515004 to B.S.A., the National Natural ScienceFoundation of China (NNSFC) [No. 81,801,207] toPS, as well as NNSFC [No. 81,671,186] to LC.

98. Longitudinal comparison of real-timeconversion and immunohistochemistry fordetection of chronic wasting disease in orallyexposed white-tailed deer

Nathaniel D. Denkersa, Davin M. Hendersona, Clare E.Hoovera,b*, Amy V. Nallsa, Erin McNultya, Candace K.Mathiasona and Edward A. Hoovera

PRION 51

aPrion Research Center, Department of Microbiology, Immunology,and Pathology, College of Veterinary Medicine and BiomedicalSciences, Colorado State University, Fort Collins, CO, USA

CONTACT Nathaniel D. Denkers [email protected]

*Present address: AstraZeneca, Waltham, NJ

ABSTRACT

Background: Chronic wasting disease (CWD) con-tinues to expand across North America and Canada,and more recently was discovered in Scandinavia.While the exact mechanism of CWD transmission hasyet to be elucidated, early diagnosis of infected animalsremains a priority in curtailing its spread. While immu-nohistochemistry (IHC) and enzyme linked immuno-sorbent assay (ELISA) remain the gold standards fordiagnosing CWD, real-time quaking induced conver-sion (RT-QuIC) is capable of detecting substantiallylower concentrations of prions, which may translateinto detection earlier in disease course. In this study,we sought to compare the sensitivity of RT-QuIC andIHC on a series of longitudinal tonsil and recto-analmucosal lymphoid tissue (RAMALT) biopsies fromwhite-tailed deer during the course of CWDprogression.

Methods: White-tailed deer were inoculated via the peros (PO) route with CWD(+) [n = 20] and CWD(-)[n = 4] inocula (brain homogenate or saliva) and pairedtonsil and RAMALT biopsies were collected every3 months thereafter. Biopsies were assayed for prionseeding activity by RT-QuIC and for PrPCWD deposi-tion by IHC.

Results: RT-QuIC detected seeding activity in 10 of 20(50%) tonsil biopsies prior to IHC detection, on averageof 5.1 months (3–15 months) earlier. In RAMALTbiopsies, detectable seeding activity was observed in13 of 20 (65%) samples prior to IHC detection, onaverage 4.6 months (3–15 months) earlier. In theremaining biopsies, detection was concurrent by bothmethods. At no sampling point did a positive IHCresult precede a positive RT-QuIC result. Of note, forbiopsy samples that had already been determined posi-tive by both RT-QuIC and IHC in a previous collection,RT-QuIC detected seeding activity in seven (7) tonsiland four (4) RAMALT follow-up biopsies even thoughno lymphoid follicles were present in the paired sam-ples to assess positivity by IHC. All biopsies from CWD(-) deer were negative by both assays throughout thestudy. Overall, these results demonstrate that RT-QuICdetected CWD positivity in both tonsil and RAMALTbiopsies approximately 5 months prior to IHC.Moreover, seeding activity was detectable even when

(due to poor biopsy sampling) lymphoid follicles werenot present to permit meaningful IHC analysis.Conclusions: These data indicate that RT-QuIC ismore sensitive than IHC in detecting CWD infection,and could thereby be useful in determining futuremanagement strategies.

Funding

Supported by NIH R01-NS-061902, P01-AI-077774,F30-ODO-118,143, T32-OD0-10,437

99. Cellulose ethers interfere with CWD prionpropagation in vitro and in vivo

Preetha Gopalakrishnana, Jie Yua, Samia Hannaouia,Sheng Chun Changa, Maria Arifina, Katsumi Doh-urab,Hermann Schatzlc and Sabine Gilcha

aDepartment of Ecosystem and Public Health, Calgary PrionResearch Unit, Faculty of Veterinary Medicine; Hotchkiss BrainInstitute; University of Calgary, Calgary, Canada; bDepartment ofNeurochemistry, Tohoku University Graduate School of Medicine,Sendai, Japan; cDepartment of Comparative Biology andExperimental Medicine, Faculty of Veterinary Medicine; HotchkissBrain Institute; University of Calgary, Canada

ABSTRACT

Chronic wasting disease (CWD) is a prion disease of freeranging and farmed cervids. CWD is highly contagious andthe absence of an effective vaccine makes a complete era-dication of CWD not realistic to date. Recently, a singlesubcutaneous injection of Cellulose Ethers (CEs) wasreported to markedly extend the life span of transgenicmice infected with 263K prions. Despite the fact that themechanisms of action of the CEs are still a matter ofdiscussion, this approach could be a good strategy againstCWD spreading, thus, limiting its potential in transmissionto cervid and non-cervid animals and potentially tohumans. In this study, we used the cellulose ethers TC-5RW and 60SH in vitro and in vivo to assess their efficacyto interfere with CWD prion propagation. In vitro, TC-5RW displays a dose dependant response to inhibit RT-QuIC reaction seeded with different CWD isolates fromwhite-tailed deer (WTD), mule deer (MD) and elk. Theinhibitory effect starts with a concentration as low as0.1 μg/ml and totally inhibits conversion at 10 μg/ml.Groups of transgenic mice overexpressing elk PrPC weresubcutaneously (s.c.) injected with the CE compounds andinoculated intracerebrally (i.c.) with different CWD isolates(elk, WTD, and MD) the same day (group A) or onemonth after the CE injection (group B). All treated groupsshow a significantly prolonged incubation period. Survivalis extended between +12% to +32% in mice of group A,and +20% to +31% in mice of group B, depending on the

52 ABSTRACT

CWD isolate used for inoculation, when compared to thecontrol group. The prolonged incubation period in thetreated groups correlates to a significantly reduced resis-tance of prions to proteinase K (PK). This is demonstratedby Western blot analyses of 10% brain homogenatedigested with a range of PK concentration. Interestingly,upon passage of brain homogenates form control andgroup B mice, tgElk mice inoculated with group Bmaterialshowed a prolonged life span of +17% without further CEtreatment, supporting the hypothesis of a modified state ofinfectivity in the mice treated during first passage. Insummary, our results demonstrate that CEs are efficientin inhibiting CWD prion propagation independent of theorigin of the isolates used for inoculation.We show that CEtreatment alters biochemical properties and reduces infec-tivity of prions in vivo. Therefore, these compounds can beuseful in limiting the spread of CWD.

100. RT-QuIC detection of pathological prionprotein in subclinical goats following experimentaloral transmission of L-type BSE

Alessandra Favolea, Maria Mazzaa, Elena VallinoCostassaa, Antonio D’Angelob, Guerino Lombardic,Claudia Palmitessaa, Dell’Atti Luanaa, Paola Crociaraa,Elena Berronea, Marina Galloa, Monica Lo Faroa,Tiziana Avanzatoa, Pier Luigi Acutisa, CristinaCasalonea and Cristiano Coronaa

aIstituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valled’Aosta; bUniversity of Turin, Department of Veterinary Science;cIstituto Zooprofilattico Sperimentale della Lombardia e dell’EmiliaRomagna

CONTACT Alessandra Favole [email protected]

ABSTRACT

In vitro assays such as serial protein misfolding amplifica-tion and real-time quaking-induced conversion (RT-QuIC) allow assessment of the conversion of the cellularprion protein (PrPC) into a pathogenic misfolded form(PrPSc). RT-QuIC can be used for the detection of typicaland atypical prions in a variety of biological tissues fromhumans and animals. It has been demonstrated that L-typeBovine Spongiform Encephalopathy (L-BSE) can be trans-mitted to a range of species including sheep1. and goats2. byintracranial inoculation. However, information on suscept-ibility of small ruminants to atypical BSE prions throughoral transmission are still lacking.

In the present work four Saanen and one mixed breedgoats were orally administered with 15ml of a 30% (wt/vol)L-type BSE brain tissue homogenate (5 g tissue). Despite aprolonged observation period of 80months, all the animalsdid not develop clinical signs referable to TSEs and tested

negative by standard diagnostics performed on brainstem.Next, pooled brain samples from the same animals wereanalyzed by western blot (WB) and RT-QuIC by usinghamster-sheep (Ha-S) and Ha 90-231 recombinantPrPSen. All the examined samples resulted negative at WBwhereas three out of five showed positive RT-QuIC reac-tions. Furthermore, prion distribution in multiple brainregions was assessed in two out of three goats resultedpositive. Preliminary results demonstrated RT-QuIC seed-ing activity in various brain regions including frontal cortexand cerebellum. Our data confirm the susceptibility of goatspecies to bovine TSEs and the applicability of RT-QuIC asa highly sensitive assay able to detect prions in subclinicalanimals. Furthermore, based on the results achieved in thisstudy, a revision or implementation of traditional methodsof diagnosis could be obtained.

References

[1] Simmons MM, et al. Vet Res. 2016;47(1):112.[2] Vallino Costassa E, et al. PLoS One. 2018;13(5):

e0198037.

101. Application of high-throughput, capillary-based Western immunoassays to modulatedcleavage of PrPC

Andrew R. Castlea,b, Nathalie Daudea, Sabine Gilchc

and David Westawaya,b,d

aCentre for Prions and Protein Folding Diseases, University ofAlberta, Edmonton, Canada; bDepartment of Medicine, Universityof Alberta, Edmonton, Canada; cDepartment of Ecosystem andPublic Health, Calgary Prion Research Unit, Faculty of VeterinaryMedicine and Hotchkiss Brain Institute, University of Calgary,Calgary, Canada; dDepartment of Biochemistry, University ofAlberta, Edmonton, Canada

CONTACT Andrew R. Castle [email protected]

ABSTRACT

The C-terminal fragment of the cellular prion protein(PrPC) that is generated physiologically by the alpha-clea-vage pathway is resistant to misfolding and acts as a domi-nant-negative inhibitor of prion replication. This fragment,known as C1, also lacks the reported binding sites forneurotoxic beta- amyloid and alpha-synuclein oligomersthat are present in full length (FL) PrPC. In contrast, a rarerproteolytic pathway – beta-cleavage – produces the longerC-terminal fragment C2 that retains the ability to misfoldand may be resistant to cleavage at the ‘alpha’ site. Thus,modulators of PrPC proteolysis that enhance alpha-clea-vage or inhibit beta-cleavage are of interest therapeutically.To identify suchmodulators, we initiated cell-based screensof small molecule libraries to identify compounds with

PRION 53

effects on PrPC proteolysis. We decided to measure thelevels of FL PrPC and its cleavage fragments using anautomated, capillary-based Western immunoassay system;this technique offers higher throughput and more accuratequantification than traditionalWestern blotting. Firstly, weoptimised the detection and quantification of FL PrPC andits cleavage fragments in cell lysate samples treated withpeptide-N-glycosidase F to remove the N-linked glycansnormally present. Secondly, we optimised the simultaneousquantification of PrPC and beta-tubulin levels to allow forloading error correction. Thirdly, we developed a protocolfor performing all cell culture, lysis and deglycoslyationsteps in 96-well microplates before capillary Western ana-lysis and demonstrated that this approach resulted in com-parable intra-assay percentage coefficients of variation tousing standard protocols for these steps. Finally, wescreened the ApexBio DiscoveryProbe Protease InhibitorLibrary by exposing RK13 cells expressing the artificial ‘S3’variant of PrPC to each compound at 20 µM concentrationfor 4 days; S3 PrPC displays accentuated beta-cleavage inRK13 cells, thereby enabling C2 levels to be quantifiedreliably. Analysis of the screening data revealed distinctclasses of inhibitors with differing effects on PrPC fragmen-tation products.

KEYWORDS: Prion disease; neurodegeneration; pro-tein misfolding; proteolysis; fragmentation; high-throughput screening; capillary Western

102. Distinctive PrP particles comprise hamster-adapted prion strains

Leonardo M. Corteza,b, Camilo Duque Velázqueza,b,Debbie McKenziea,c, Valerie L. Sima,b

aCentre for Prions and Protein Folding Diseases, University ofAlberta; bDepartment of Medicine, Faculty of Medicine &Dentistry, University of Alberta; cDepartment of BiologicalSciences, Faculty of Science, University of Alberta

CONTACT Leonardo M. Cortez [email protected]

ABSTRACT

Background: The conversion of PrPC into a pathogenicconformation results in multimeric PrPSc particles thatrange in size from small detergent-soluble oligomers tolarge insoluble aggregates. The prion strain phenomenon,characterized by different phenotypes/pathologies of dis-ease, may be explained by PrPSc conformational differencesresulting in reproducible strain-specific PrPSc multimericstates. However, the complexity, variability, and propertiesof these PrPSc particle distributions are not well established.It has been hypothesized that prion strains are comprised ofa cloud of PrPSc conformations with different aggregationproperties. We present a detailed study of fractionated

PrPSc particles to better understand prion strains, transmis-sion barriers, drug resistance, and therapeutic targets.Methods: Asymmetric-flow field-flow fractionation (AF4),a one-phase chromatography technique of size-basedseparation, was used to isolate soluble prion particlesfrom brain homogenate of 263K, Hyper (HY) andDrowsy (DY) infected hamsters. We determined thehydrodynamic radius (Rh) and amount of total and PK-resistant PrP in each fraction using dynamic light scatteringand immunoblotting. Using different AF4 buffer condi-tions, we studied the stability of different PrP oligomers.Particle size was correlated with RT-QuIC seeding activity.Results: Each strain generated a reproducible distributionof PrP particle sizes. For HY and 263K, there was a popula-tion of smaller PrP particles (Rh = 2-8nm) that weresensitive to PK digestion but had strong seeding activity.A second population of larger PrP particles (Rh~15-200nm) was detergent and PK resistant but had less seed-ing activity than the smaller particles. In contrast, DY hadpredominantly a smaller population of PrP particles, peak-ing at Rh~7nm. These smaller DY particles were sensitiveto PK but had less seeding activity than 263K and HY. Thefew large DY PrP particles were resistant to PK but sensi-tive to detergent, and their seeding activity was strongerthan large HY and 263K PrP particles.

Interestingly, the size distribution of PK-resistant PrPparticles was almost identical for all strains, with PK resis-tance being evident in particles with Rh>20nm. This sug-gests that PK resistance is related to the size of PrP particle.Conclusions: The distinct PrPSc particle size distributionand oligomer properties in different strains suggest thereare mechanisms to maintain a stable PrPSc population ineach strain. Hyper and 263K, which have similar diseaseoutcomes, are comprised of similar distributions of PrPSc

particles; Drowsy, with very a different disease outcome,has different amounts of aggregates with different bio-chemical properties. PK sensitive PrP appears to play acentral role in these differences.

103. Biochemical comparison of peripheral vs. centralnervous system-derived PrPCWD in white-tailed deer

Lindsay E. Parrie, Davin M. Henderson, Clare Hoover,Kristen A. Davenport, Sarah K. Cooper and Edward A.Hoover

Prion Research Center, Colorado State University, Fort Collins,CO, USA

CONTACT Lindsay E. Parrie [email protected]

ABSTRACT

In nature, cervids are infected with chronic wasting disease(CWD) by oral and nasal mucosal exposure. However,PrPC expression is not the primary driver of PrPCWD

54 ABSTRACT

accumulation, as tissues with the highest PrPC expressionare not the first to accumulate PrPCWD. Studies of earlyCWD pathogenesis have implicated pharyngeal lymphoidtissues as the earliest sites of prion accumulation. PrPCWD

replication differs between peripheral tissues and the CNS.PrPCWD deposits in lymphoid tissues are smaller and morediffuse than those found in brain, which is typically char-acterized by large amyloid plaques. Additionally, lym-phoid-derived PrPCWD appears to be more sensitive toproteinase K digestion than is brain PrPCWD, despiteequivalent seeding activity as measured in RT-QuIC. Ourstudies address two main questions: (1) Do lymphoidprions have different biophysical properties than thosefrom the CNS? (2) Do lymphoid prions have differentinfectivity properties than CNS prions? To investigate thepotential differences between peripheral PrPCWD andprions formed in the CNS, we orally exposed white-taileddeer (WTD) to very lowdoses of infectiousCWD+saliva orbrain homogenate. Tissueswere collected at clinical disease,and prions from retropharyngeal lymph node and obexcompared as to biophysical properties, conversion seedingefficiency, protease sensitivity, and immuno-blotting pro-files. To functionally assess infectivity of peripheral andCNS-derived PrPCWD, we will perform both cervid prioncell assay (CPCA) and bioassay of cervidized transgenicmice via oral inoculation. These studies will illuminatepreviously unknown features of CWD peripheral prionpathogenesis that likely relate to the efficient horizontaltransmission of CWD.

KEYWORDS: Chronic wasting disease; lymph node;RT-QuIC

104. Development of a high-throughput assay fortau purification and aggregation

Allan Yarahmady and Sue-Ann Mok

Department of Biochemistry, University of Alberta, Edmonton,Canada

CONTACT Allan Yarahmady [email protected]

ABSTRACT

Introduction: To study the process of tau aggregation,researchers have commonly utilized in vitro experimentswhere tau expressed and purified from E. coli is aggregatedvia addition of inducers such as heparin. However, thestructures of artificially obtained tau fibrils differ fromthose found in actual Alzheimer’s disease patients.Differences in structures suggest that the folding pathwaysof in vitro aggregation differ from those in vivo. Thesedifferences call into question the relevance of in vitroaggregation models. Development of potential treatments

that inhibit or reverse tau aggregation would likely be aidedby in vitro derived fibrils that more closely mimic thestructures observed in Alzheimer’s patients. Point-muta-tions of tau have been shown to significantly alter the corestructure of the generated fibrils. New methodologies suchas deep mutational scanning of the tau sequence couldidentify specific mutants that give a fibril core structureidentical to that of patient-derived fibrils. In order to per-form a large-scale screen of tau mutants, methods for high-throughput expression, purification, and aggregate analysisof tau were optimized.Materials and Methods: Tau was expressed in three milli-litre cultures in 48-well deep plates. The pellets were lysedwith lysozyme andboiled to denature contaminant proteinsand cell debris. The lysates were spun down and the super-natants were isolated which contain soluble tau. Thekinetics of tau aggregation were studied via a thioflavin Ttime-course assay using heparin as an inducer. Core struc-tures of the aggregates from the kinetic assays were char-acterized using trypsin digestion and visualization by SDS-PAGE.Results: It was found that tau was readily expressed andpurified in small-scale. The yield from high-throughputsmall-scale expression of tau is consistent and the aggrega-tion propensity of tau is able to be studied using a low-volume thioflavin T kinetic assay. The trypsin-resistantcore of the fibrils derived from this assay resembles thoseobtained through traditional purification and in vitroaggregation techniques.Conclusions: It is possible to obtain recombinantlyexpressed tau of high yield and purity using small-scalemethods. The aggregation kinetics and fibril core structuresof these tau extracts are also able to be characterized bythioflavin T assay and trypsin digestion, respectively. Assuch, a high-throughput deep mutational screen of tau ispossible and can be used to test if the tau fibril structuresobserved in Alzheimer’s can be recapitulated in vitro.

105. Differential binding of prions to vegetation

Elizabeth Triscotta, Alsu Kuznetsovab, DebbieMcKenziea and Judd Aikenc

aCentre for Prions and Protein Folding Diseases, Department ofBiological Sciences, University of Alberta; bDepartment of RenewableResources, University of Alberta; cCentre for Prions and Protein FoldingDiseases, Department of Agriculture Food and Nutritional Sciences,University of Alberta

CONTACT Elizabeth Triscott [email protected]

ABSTRACT

Chronic wasting disease (CWD), a prion disease ofcervids, is present in North America and Europe. Itaffects a variety of captive and free ranging animals,

PRION 55

including mule deer, white tailed deer, reindeer, moose,and elk. Preclinical and clinical animals shed infectivityin a number of bodily fluids, including saliva, faeces,and nasal secretions. These prions are resistant todegradation and can contaminate the environment formany years. Factors such as the surface composition ofmaterials and the mineralogy of soils can affect whereinfectivity accumulates in the environment. Given,however, that in most environments prions will initiallycontact vegetation, understanding the factors that affectthe interaction between prion infectivity and vegetationis of utmost importance. We developed an assay inwhich infectious brain homogenate is applied to vege-tation samples, which are then rinsed. The proteins areprecipitated from the rinse water and the amount ofPrP that rinses off each vegetation sample was com-pared. Given that surface composition affects prionadherence, we first compared lichen, which is coatedwith polysaccharides, and grass, which is covered with awaxy cuticle. Our data indicate that less prions rinse offlichen than grass. Furthermore, the binding of CWDprions to a variety of plants shows that the prion-vegetation interaction is affected by properties of theplant, e.g. microarchitecture and precise chemical com-position. Freezing and drying of the plants alters thisinteraction. These data give us insight in how infectivitymoves through the environment and has ramificationsfor the plausibility of environmental CWD transmis-sion. Understanding where prions accumulate will helpin developing methods for environmental monitoringand decontamination.

106. Long-read nanopore sequencing provides fastand accurate identification of genetic variants inthe human PRNP gene

Dimitriadis Athanasios, Francois Kroll, Tracy Campbell,John Collinge, Simon Mead, Emmanuelle Viré andJohn Collinge

MRC Prion Unit at UCL, Institute of Prion Diseases, Queen SquareHouse, Queen Square, University College London, London, UK

CONTACT Dimitriadis Athanasios [email protected]

ABSTRACT

The human prion gene (PRNP) is a 15.5 kb sequence onchromosome 20 which encodes the prion protein (PrP). Itis composed of two exons flanking a single large intron.The protein coding region is a 762-bp region within thesecond exon. Genetic variants identified in the PRNP geneare associated with prion diseases, such as Creutzfeldt-Jakob disease (CJD). Previous work has demonstrated

that the number of OctaPeptide Repeats (OPRs) in thegene coding region is in general inversely correlated withage of disease onset [1]. Similarly, it is well established thatgenotype of the codon 129 strongly affects disease onset,severity, and phenotype [2]. This evidence highlights thenecessity of accurate genetic testing in CJD management.Here we tested how long-read sequencing technologiescompare to the well-established Sanger sequencingmethod, using a Nanopore MinION sequencer. First, wesequenced full-length PRNP in 9 individuals known to becarrying various types ofOPR insertions (OPRIs), deletions(OPRDs), and SNVs (E200K, P102L) and confirmed theirgenotype. To process this data, we designed a bioinfor-matics pipeline based on minimap2, sniffles, nanopolish,and customR scripts to automate the analysis and filter ourresults. Then, we sequenced the full-length prion gene in 10patients with sporadic CJD and characterized polymorph-isms in the intronic region and the promoter of the gene.Altogether our results reveal that long-read sequencing,using MinION, allows investigating the genetic makeupof the whole PRNP in patients, including the promoter,flanking regions, intron, and both exons of the gene. Wepropose an accurate, quick, and user friendly method thatbypasses the needs for expensive instruments and whichallows for sample multiplexing. Finally, our pipeline is alsoable to report the results in an easily readable text format.

KEYWORDS:Nanopore sequencing; genetic prion disease; bioinfor-matics; structural variants; single-nucleotide variations

References

[1] Brown K, et al. J Geriatr Psychiatry Neurol. 2010;23(4):277–298.

[2] Schmitz M, et al. Mol Neurobiol. 2017;54(6):4138–4149.

107. Global analysis of protein degradation ratesin prion infected cells by dynamic SILAC

Charles R. Hutti, Kevin A. Welle, Jennifer R.Hryhorenko and Sina Ghaemmaghami

University of Rochester, New York

CONTACT Charles R. Hutti [email protected]

ABSTRACT

Prions diseases are rare neurological disorderscaused by the misfolding of cellular prion protein(PrPC). The misfolded conformers aggregate intocytotoxic fibrils (PrPSc) that facilitate the conver-sion of additional prion proteins into the misfoldedform. Intracellular PrPSc aggregates primarily

56 ABSTRACT

accumulate within late endosomes and lysosomes,organelles that participate in the degradation andturnover of a large subset of the proteome. Thus,intracellular accumulation of PrPSc aggregates mayglobally disrupt protein degradation kinetics withininfected cells. To test this hypothesis, we analyasedthe effect of prion infection on protein degradationrates in N2a cells using dynamic stable isotopiclabelling with amino acids in cell culture (dSILAC) andbottom-up proteomics. We were able to analyse thedegradation rates of ~5000 proteins, constituting the lar-gest proteome-wide analysis of degradation rates in prioninfected cells to date. As expected, the data indicate thatthe degradation rate of the prion protein is significantlydecreased in infected cells. Indeed, we demonstrate thatcellular dilution due to cell division (rather than degrada-tion) is the dominant factor in clearance of PrPSc ininfected N2a cells. Conversely, the degradation kineticsof the remainder of the N2a proteome generally increasesupon infection. This effect occurs concurrently withenhancements in cellular activities of the proteasomeand lysosomal hydrolases. Our data indicate that N2acells respond to prion infection by globally upregulatingthe cellular protein degradation machinery. The resultingenhancement in proteome flux may play a role in thesurvival of N2a cells in the presence of prion infection.

108. Crystal structural studies of Fab fragmentsbound to the prion molecule and perspectives onthe loop connecting β2 to α2

Pravas Kumar Barala, Mridula Swayampakulaa,Adriano Aguzzib and Michael N. G. Jamesa

aDepartment of Biochemistry, Faculty of Medicine and Dentistry,University of Alberta, Edmonton, Canada; bDepartment ofPathology, Institute of Neuropathology, University Hospital Zurich,Zurich, Switzerland

CONTACT Michael N. G. James [email protected]

ABSTRACT

Our laboratories have collaborated on determining thestructures of the globular domains of prion moleculesfrom several mammalian species by X-ray crystallogra-phy. We have used the advantage of co-crystallizing thefolded domains of the cellular prion proteins (PrPC) withthe Fab fragments of several immunoglobulins raisedagainst the mouse prion. In particular, the Fab fragmentsof POM1 and POM6 have adjacent but slightly differentbinding epitopes on the mouse PrPC globular domain.Biological studies on these antibodies are quite interest-ing; POM1 is a highly toxic antibody whereas POM6 is aninnocuous antibody. We have determined the structures

of POM1 Fab bound to the folded domains of the PrPC sof mouse, human, bovine, deer, elk, and Syrian hamster.The structure of mouse PrPC bound to the Fab fragmentof the POM6 antibody has also been determined.We havecompared the loop connecting the β2 strand to the α2helix of the PrPC molecules. In all structures determinedby X-ray crystallography, this loop adopts very similarstructures involving a 310 helix and the α2 helix initiatorresidue Asn171. Another very important residue in thisloop is Tyr169 and its interactions with residues on helixα2. We have compared the structures of this loop deter-mined by X-ray crystallography to the structures of thisloop as determined by NMR spectroscopy. This regiondiffers in conformation by a substantial amount in thestructures determined by the two techniques (the Cαatoms of the two Ser170 residues differ in position by5.1Å between the X-ray structure and the NMR struc-ture). Tighter contact between the residues of the β2-α2loop and residues from the β1- β2 region seems to be thekey in keeping the folded prion domain more stable.

109. Analysis of N-linked glycans of ovine prionprotein by liquid chromatography coupled withelectrospray mass spectrometry

Natali Nakića, Thanh Hoa Trana, Mislav Novokmetb,Jelena Šimunovićb, Olivier Andreolettic, GordanLaucb and Giuseppe Legnamea

aLaboratory of Prion Biology, Department of Neuroscience, ScuolaInternazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy; bGenosGlycoscience Research Laboratory, Zagreb, Croatia; cUMR INRA ENVT1225- IHAP, École Nationale Vétérinaire de Toulouse, Toulouse, France

CONTACT Natali Nakić [email protected] Laboratory of PrionBiology, Department of Neuroscience, Scuola InternazionaleSuperiore di Studi Avanzati (SISSA), Trieste, Italy

ABSTRACT

Prion diseases are characterized by a distinct pathology inthe central nervous system, however, differing from oneanother in clinical symptoms, incubation time, biochem-ical properties, and in the deposition pattern. These differ-ences arise from the fact that PrPC can acquire multipleconformationally different PrPSc states, called strains. Thequestion of how different strains display such a diversity indisease phenotypes remains to be answered. One of theways strains are characterized is by immunoblotting, whereeach strain shows a specific three band pattern. The reasonfor this comes from the fact that the prion protein can carryup to two N-linked glycans. The knowledge about inter-individual or inter prion strain differences in N-glycanstructures is still not fully understood. The aim of thisstudy is to differentiate individual N-glycan structures ondifferent sites and to evaluate functional aspects of

PRION 57

variation regarding prion protein glycosylation. The analy-sis was performed on VRQ/VRQ sheep brain affected withclassical scrapie. The ovine PrPSc was isolated and purifiedby immunopurification and SDS-PAGE. The isolation ofthe scrapie prion protein was shown to be somewhatchallenging, since glycoproteins are heterogeneously glyco-sylated, this leads to a decreased concentration of eachanalyte (each specific glycan). After obtaining a sufficientamount and purifying the protein, the di- and monoglyco-sylated forms of the prion protein were extracted from thegel and in-gel trypsin digestion was performed. The trypticglycopeptides were then analysed using reverse phaseliquid chromatography coupled with electrospray massspectrometry (LC-ESI-MS). The glycan structures foundon the ovine PrPSc have been shown to contain ‘brain-specific’ N-glycans, which are high in fucose and sialicacids. Other features found, that are characteristic forbrain N-glycans, include hybrid-type structures, bisectingN-Acetylglucosamine (GlcNAc) and core (α1-6)fucose, aswell as outer arm (α1-3)fucose. We were able to develop amethod for isolation of ovine PrPSc from infected tissueand analysing the glycopeptides with LC-ESI-MS. Withthis approach we plan to compare glycan structuresbetween different ovine prion strains in order to betterunderstand whether or not N-glycans contribute to prionstrain differences.

KEYWORDS: Prion; N-glycan; LC-ESI-MS

110. Involvement of MAPK pathway and tau in pα-syn*-induced mitotoxicity

Diego Grassia,b, Natalia Diaz-Perezc, Laura A.Volpicelli-Daleyd and Corinne Ida Lasmézasa,b

aDepartment of Immunology and Microbiology, The ScrippsResearch Institute, Scripps Florida, Jupiter, FL, USA; bDepartmentof Neuroscience, The Scripps Research Institute, Scripps Florida,Jupiter, FL, USA; cPrivate address, Palm Beach Gardens, FL, USA;dCenter for Neurodegeneration and Experimental Therapeutics,University of Alabama at Birmingham, Birmingham, USA

CONTACT Corinne Ida Lasmézas [email protected]

ABSTRACT

We recently identified a phosphorylated form of α-synu-clein, pα-syn*, as a key neurotoxic α-synuclein speciesfound in cultured neurons, as well as in mouse andParkinson’s disease patients’ brains. Small pα-syn* aggre-gates localize to mitochondria and induce mitochondrialdamage and fragmentation. Herein, we investigated themolecular basis of pα-syn*-induced toxicity. By immuno-fluorescence, we found phosphorylated MKK4, JNK,ERK5 and p38 MAPKs in pα-syn* inclusions. pJNK colo-calized with pα-syn* at mitochondria and mitochondria-

associated ER membranes where it was associated withBiP and pACC1. We also found that pα-syn* aggregatesare tightly associated with small ptau aggregates of similarsize. Pα-syn*/ptau inclusions localized to areas of mito-chondrial damage and to mitophagic vesicles, showingtheir role in mitochondrial toxicity, mitophagy induction,and their removal along with damaged mitochondrialfragments. Several MAPKsmay act cooperatively to phos-phorylate tau, notably JNK, p38, and GSK3β, a non-MAPK that was also found phosphorylated in the vicinityof pα-syn*/ptau aggregates. Thus, pα-syn* appears to bethe trigger of a series of kinase mediated pathogenicevents and a link between α-syn pathology and tau,another protein known to aggregate in Parkinson’s dis-ease and other synucleinopathies.

KEYWORDS: Alpha-synuclein; tau; Parkinson’s dis-ease; primary neurons; kinases; neurotoxicity; mito-chondria; mitophagy

111. Identification of rare genetic variantsassociated with sporadic CJD by exome sequencing

Helen Speedya, Penny Norsworthya, Holger Hummericha,Gary Adamsona, James Uphilla, John Collingea andSimon Meada

aMRC Prion Unit at UCL, UCL Institute of Prion Diseases, CourtauldBuilding, London, UK

CONTACT Helen Speedy [email protected]

ABSTRACT

Background: Prion diseases are progressive neurodegen-erative conditions characterized by the misfolding andaggregation of prion protein. Sporadic Creutzfeldt-JakobDisease (sCJD) is themost common human prion disease.The common polymorphism at codon 129 of the PRNPgene influences sCJD risk; however additional geneticfactors are thought to contribute to disease susceptibility.Ongoing genome-wide association studies have identifiedcommon genetic variants associated with sCJD risk. Inthis study we use exome sequencing to investigate the roleof rare, disruptive variants as determinants of susceptibil-ity to sCJD.Materials and Methods: This study incorporates exomesequencing of an in-house set of 249 sCJD cases as well asdata from 126 sCJD cases from the Medical ResearchCouncil (MRC) Brain Bank Network. Controls arederived from publically available resources.

In-house, genomic DNA was extracted from peripheralblood. Exome capture using Agilent technology andpaired-end sequencing on an Illumina HiSeq2000 sequen-cer was performed at Source Bioscience (Nottingham,UK).

58 ABSTRACT

Sequencing reads were aligned to the human referencegenome (hg19) using Burrows-Wheeler Aligner software.Alignments were processed using the Genome AnalysisTool Kit pipeline, according to best practices. Variantswere filtered according to quality and frequency metrics,as well as predicted functional consequences. Single variantand gene-based association testing was performed.Results: Initial analyses of exome sequencing data failed todetect a single rare, disruptive genetic variant significantlyassociated with sCJD risk, whilst no individual gene inburden testing surpassed genome-wide significance.Future work will focus on exploring alternative rare variantanalysis methods and the subsequent prioritization of var-iants for validation and follow-up in additional sCJD cases.In conjunction, we will examine the burden of clinicallyrelevant (novel or known pathogenic) mutations in knowndementia genes, deriving candidate genes from the MRCDementia Gene Panel, UK Genetic Testing NetworkClinical Panels and through manual literature curation.Conclusions: We have conducted an exome sequencingstudy involving 375 sCJD cases to examine the role of rare,disruptive genetic variants in sCJD risk. Although no singlevariant or gene of significant impact was discovered ininitial analyses, further exploration of this dataset isongoing.

KEYWORDS: Genetics; exome; rare variant; sporadicCreutzfeldt-Jakob disease

112. The low levels of nerve growth factor and itsupstream regulatory kinases in prion infection isreversed by resveratrol

Chao Hua,b*, Cao Chenb,c*, Jia Chena, Kang Xiaob,c,Jing Wangb,c, Qi Shib,c, Yue Mab, Li-Ping Gaob, Yue-Zhang Wub, Lian Liua, Ying Xiaa, Pu Yana, AdalaitiMaimaitimingb, Dong-Hua Zhoub, Li-Na Zhangb,Zhi-Bao Chena and Xiao-Ping Dongb,c,d

aCollege of Life Science and Technology, Heilongjiang BayiAgricultural University, Daqing, China; bState Key Laboratoryfor Infectious Disease Prevention and Control, NationalInstitute for Viral Disease Control and Prevention, ChineseCenter for Disease Control and Prevention, Beijing, China;cCollaborative Innovation Center for Diagnosis and Treatmentof Infectious Diseases, Zhejiang University, China; dCenter forGlobal Public Health, Chinese Center for Disease Control andPrevention, Beijing, China

CONTACT C. Chen [email protected]; Z. B. Chen [email protected]; X. P. Dong [email protected]

* These authors contributes equally to this work.

ABSTRACT

Up to the present, none of the specific vaccines ortherapeutic drugs has been documented to be

effective in human or animal prion diseases.Recently, resveratrol shows the ability to eliminateprion replication. To obtain insights into the possi-ble changes of brain neurotrophic factor (NGF) andits upstream regulatory cascade during prion infec-tion and after removal of prion propagation, thelevels of NGF and its upstream regulatory factorsin various SMB cell lines and in the mice brainsinoculated with various SMB cellular lysates wereassessed with various methodologies. The levels ofNGF were significantly decreased in vivo and invitro, while recovered after removal of PrPSc byresveratrol. Morphological assays revealed that theNGF signals mainly colocalized within neurons, butnot in the proliferative astrocytes and microglia. Theupstream positive regulatory kinases, such asp-CREB, p-CaMKIV, CaMKK2 were also remark-ably decreased in the prion infected cells and micebrains, whereas the negative regulatory one,p-CaMKK2, was increased. The aberrant situationsof those kinases in prion infected cell lines or micebrains could be also partially reversed by removal ofprion agent. Moreover, we demonstrated that thesignals of CaMKK2 and p-CaMKK2 were also dis-tributed predominately in neurons in the brain tis-sues. The data here illustrate a direct linkage ofabnormally repressive NGF and its upstream regu-latory kinases with prion infection. Resveratrol hasnot only the ability to inhibit prion replication, butalso to improve the expression of NGF viaCaMKK2/CaMKIV cascade, which might benefitthe microenvironment in brains.

KEYWORDS: Prion; resveratrol; NGF; CaMKIV;CaMKK2

113. Diagnosis of CWD in a herd of farmed reddeer

Ines Walther, Antanas Staskevicius, Andrei Soutyrineand Gordon Mitchell

National & OIE Reference Laboratory for Scrapie and CWD,Canadian Food Inspection Agency, Ottawa, ON, Canada

CONTACT Gordon Mitchell [email protected]

ABSTRACT

Chronic wasting disease (CWD) continues to adverselyimpact wild and farmed cervids of North America,affecting primarily white-tailed deer (Odocoileus virginia-nus), mule deer (Odocoileus hemionus), and elk (Cervuscanadensis). Red deer (Cervus elaphus) are closely relatedto elk, and occasional cases of CWD have been reported

PRION 59

in farmed red deer in the United States and the Republicof Korea, and in a wild red deer in Norway. CWD wasrecently detected in farmed red deer for the first time inCanada, in a region geographically distinct from all pre-vious cases of CWD. Understanding the diagnostic fea-tures and pathogenesis of CWD in red deer is essentialto developing effective disease detection and controlstrategies in this species. The index case of this herd, aclinically healthy 15-month old male red deer, was initi-ally detected during routine slaughter surveillance testingby ELISA. Confirmatory testing by immunohistochemis-try (IHC) demonstrated modest aggregates of pathologi-cal prion protein (PrPCWD) in restricted regions of theobex of the medulla and retropharyngeal lymph nodes.Western blot of the obex homogenate revealed a bandingpattern consistent with that of other Canadian CWDcases, and distinct from what has been observed in reddeer experimentally infected with bovine spongiformencephalopathy. Subsequent disease control measures inthis herd resulted in the testing of over 1700 red deerand an additional 10 cases of CWD were identified infemales ranging in age from 18 to 28 months. All caseswere positive in the obex by ELISA, IHC, and Westernblot, although the extent of PrPCWD deposition detectedby IHC was somewhat variable. A higher degree ofvariability in PrPCWD accumulation was found in lym-phoid tissues, with PrPCWD not detectable by IHC in thetonsils of several cases. Sequencing of the prion proteingene from all positive cases and a subset of negative casesidentified variability at several codons (98, 168, and 226)although the extent to which these polymorphisms mayinfluence susceptibility to infection is not yet evident.Natural transmission of CWD occurs relatively efficientlyamongst cervids, facilitating the geographical dissemina-tion of disease to previously naive populations.Determining the unique characteristics of CWD in differ-ent cervid species is needed to inform the refinement ofdisease management approaches.

114. Structural analysis of human and transgenicmouse-derived tau protein aggregates

Shelaine C. Flecka,b, Ghazaleh Eskandari-Sedighia,b,Razieh Kamali-Jamila,b, Mirka Kasirovac, Chae Kimc,Howard Youngb, David Westawaya,b,d, Jiri Safarc andHolger Willea,b

aCentre for Prions and Protein Folding Diseases, University ofAlberta, Edmonton, AB, Canada; bDepartment of Biochemistry,University of Alberta, Edmonton, AB, Canada; cDepartments ofPathology and Neurology, Case Western Reserve University,Cleveland, OH, USA; dDepartment of Medicine, University ofAlberta, Edmonton, AB, Canada

CONTACT Shelaine C. Fleck [email protected]

ABSTRACT

Tauopathies are a class of neurological disorders associatedwith the aggregation of the tau protein into neurofibrillarytangles. The most prominent tauopathy is Alzheimer’sdisease (AD), which presents as two forms: early onset(genetic, gAD) and late onset (sporadic, sAD). sAD doesnot have a known cause; although environmental, lifestyle,and genetic factors are thought to contribute to the diseasemanifestation. gAD can be caused by one of three differentsingle gene mutations: in the amyloid precursor protein(APP), presenilin 1, or presenilin 2. In both forms of AD,the tau protein aggregates to form intracellular fibrillarytangles, which spread along neuronal networks. Amyloid-β, the product of APP proteolysis, also contributes to ADthrough the formation of extracellular amyloid plaques.Together, these proteins lead to the extensive neuronaldeath seen in AD. The structure of tau fibrils in AD, eitheras Paired Helical Filaments (PHFs) or Straight Filaments(SFs), was solved using cryo-electron microscopy and 3D-reconstructions.1 By analysing PHFs and SFs from sAD,researchers found that filament cores consist of two iden-tical protofilaments which adopt a cross-β structure [1].Later, researchers analysed additional samples from sADand gAD cases, which confirmed the initial findings [2].Other tauopathies, including Pick’s disease and frontotem-poral dementia with parkinsonism linked to chromosome17 (FTDP-17), are caused solely by the misfolding of thetau protein. Each of these diseases is characterized by thedementia they cause and notable brain atrophy. The struc-ture of tau fibrils in Pick’s disease accumulating predomi-nantly three-repeat tau differs from AD, it forms a singleprotofilament with nine beta-strands [3]. The structure oftau fibrils in FTDP-17 is currently unknown. The focus ofour research is to further investigate the different confor-mations of the tau protein in these diseases. Using EM and3D-reconstructions we plan to examine samples from gADand sAD cases. Currently, we have examined samples fromrapidly progressive AD and a transgenicmouse line expres-sing four-repeat human tau proteinwith a P301Lmutation,which is found in some FTDP-17 patients [4]. Previously,three tau fibril morphologies: straight, coiled, and twisted-ribbon, were identified [4]. Through continued examina-tion of these fibrils using 3D-reconstructions and volu-metric visualization, we hope to see more detaileddifferences in the structure adopted by the tau protein.This will provide insights in to the role the tau proteinplays in these tauopathies and how the structural differ-ences may contribute to the distinct disease manifestations.

References

[1] Fitzpatrick, et al. Nature. 2017;547:185–190.

60 ABSTRACT

[2] Falcon, et al. Acta Neuropathol. 2018;136:699–708.[3] Falcon, et al. Nature. 2018;561:137–140.[4] Eskandari-Sedighi, et al. Mol Neurodegener.

2017;12:72.

115. Diversity of chronic wasting disease prionstrains

Camilo Duque Velásqueza, Elizabeth Triscotta, ChiyeKima, Jacques Van der Merwea, Samia Hannaouib,Trent Bollingerc, Christina Carlsond, Sylvie Benestade,Sabine Gilchb, Judd Aikenf and Debbie McKenziea

aDepartment of Biological Sciences, Centre for Prions and ProteinFolding Diseases, University of Alberta, Edmonton, Canada;bDepartment of Ecosystem and Public Health, Calgary PrionResearch Unit, University of Calgary, Calgary, Canada;cDepartment of Veterinary Pathology, Canadian Wildlife HealthCooperative, University of Saskatchewan, Saskatoon, Canada; dU.S.Geological Survey-National Wildlife Health Center, Madison, WI,USA; eNorwegian Veterinary Institute, Oslo and Trondheim,Norway; fDepartment of Agricultural, Food and NutritionalSciences, Centre for Prions and Protein Folding Diseases,University of Alberta, Edmonton, Canada

ABSTRACT

Chronic Wasting Disease (CWD) prions affect a varietyof cervid species. The expansion of the CWD geo-graphic range and the increasing prevalence makesCWD a concern for wildlife, livestock, and humanhealth. Prion pathogenesis results from the template-directed misfolding of cellular prion proteins (PrPC)into conformational species (e.g. PrPCWD) associatedwith distinct strains. We propose that cervid cellularprion protein (PrPC) polymorphisms have resulted inprion conformational diversification and speciation (i.e.by adaptive radiation) resulting in emergence of novelCWD strains. New prion strain conformers emergefollowing transmission between cervids expressing dif-ferent PrPC amino acid polymorphisms [1-4].Emergent CWD strains can have novel transmissionproperties that enable them to infect host species pre-viously considered resistant, suggesting an increase inzoonotic risk as strain-conformers diversify and evolve[2, 3]. We are comparing field CWD isolates of differ-ent cervid species from various regions of NorthAmerica and Norway and have identified differencesin biochemical properties of PrPCWD, in vitro and exvivo propagation and transmission into transgenic miceexpressing deer and elk PrP (tgDeer – 96G, tgDeer –96S, and tgElk-E226) as well as C57Bl6 mice and ham-sters. Variations in PrP-res type and protease sensitivitywere observed following treatment with proteinase K.Comparison of the PMCA seeding activity using deerand elk PrPC as substrates revealed differences betweenelk CWD isolates. ElK21 cells also responded

differently to various cervid prions. Transmission wasefficient for all isolates in tg33 and tgElk mice; however,only a few isolates were able to propagate in hostexpressing S96 deer PrPC, which has been shown toimpose a strong transmission barrier, providing ameans of differential selection of CWD strains [2].Transmission differences were observed followinginterspecies transmission. While some white-taileddeer and mule deer isolates failed to transmit intohamsters, other isolates transmitted with a low attackrate but considerable sub-clinical infection. These iso-lates had a migration pattern similar to the hamster-passaged Wisc-1 strain. Two other isolates, transmittedmore efficiently into hamsters and produced two dif-ferent PrP-res migration profiles compared to Wisc-1-like PrP-res. Transmission of Norwegian moose andreindeer CWD isolates is ongoing; preliminary resultswill be presented. Our data indicate the existence of atleast five different CWD strains based on transmissionproperties.

References

[1] Johnson CJ, et al. Prion protein polymorphisms affectchronic wasting disease progression. PloS ONE. 2011;6(3):e17450.

[2] Duque Velasquez C, et al. Deer prion proteins modu-late the emergence and adaptation of chronic wastingdisease strains. J Virol. 2015;89(24):12,362–12,373.

[3] Herbst A, et al. Chronic wasting disease prion strainemergence and host range expansion. Emerging InfectDis. 2017;23(9):1598–1600.

[4] Hannaoui S, et al. Destabilizing polymorphism in cer-vid prion protein hydrophobic core determines prionconformation and conversion efficiency. PloS Pathog.2018;13(8):e1006553.

116. Characterization of distinct SOD1 prionstrains

Jacob I. Ayersa,b, Kristy Dillona, Jeffrey Diamonda, EricFagerlia, Susan Fromholta and David R. BorcheltaaDepartment of Neuroscience, Center for Translational Research inNeurodegenerative Disease, University of Florida, Gainesville, FL, USA;bDepartment of Neurology, Institute for Neurodegenerative Diseases,University of California San Francisco, San Francisco, CA, USA

CONTACT Jacob I. Ayers [email protected]

ABSTRACT

Mutations in the Cu-Zn superoxide dismutase 1(SOD1) gene are known to be causative in 10–20% offamilial amyotrophic lateral sclerosis (fALS) cases. Theduration of disease in these patients can vary, such thatsome mutations are associated with disease of less than

PRION 61

3 years, whereas others are associated with disease ofmore than 20 years. One potential explanation for thisvariability in disease progression is that distinct SOD1mutations exhibit conformational strain properties thatmay affect their ability to propagate within the CNS. Toaddress this, we tested various preparations containingdifferent SOD1 mutations in in vivo and ex vivo seedingassays to investigate several SOD1 properties. Weinjected multiple misfolded SOD1-containing prepara-tions within the spinal cords of mice at postnatal dayP0. These recipient mice express low levels of the G85Rvariant of human SOD1 fused to YFP (G85R-SOD1:YFP) and are permissive for motor neuron diseaseinduction. Once MND was induced, the spinal cordswere analysed for the presence and morphology of theinduced G85R-SOD1:YFP aggregates by direct fluores-cence. Additionally, as an ex vivo assay, these SOD1-containing preparations were added to organotypicspinal cord slice cultures prepared from G85R-SOD1:YFP mice to directly visualize the induced aggregatesover time. We observed both distinct SOD1:YFP aggre-gate pathologies and incubation periods that bred trueupon repeated passages. Additionally, we observed thesame SOD1:YFP aggregate structures in spinal cordslice cultures as was seen in mice, when treated with agiven SOD1 containing preparation. The distinctSOD1:YFP pathologies observed suggest prion strain-like properties for SOD1. Additionally, the fact that themorphology of these structures bred true upon secondpassage in mice with the same genotype suggests thatthe formation of these aggregates are produced byconformational templated misfolding, which is respon-sible for the propagation of prions. These resultsstrongly suggest SOD1 prions are capable of exhibitingconformational strain-like variability in the misfoldingand propagation of SOD1-linked ALS.

117. Bank vole bioassay for detection of prioninfection in CWD-challenged cynomolgus macaques

Karla A. Schwenkea, Joo-Hee Waelzleina, Hermann M.Schatzlb, c, Walter Schulz-Schaeffere, Christiane Stahl-Hennigf, Stefanie Czubc, d and Michael Beekesa

aRobert Koch Institute, Prion and Prionoid Research Unit, ZBS 6, Berlin,Germany; bUniversity of Calgary, Calgary Prion Research Unit, Calgary,Canada; cUniversity of Calgary, Faculty of Veterinary Medicine, Calgary,Canada; dCanadian Food Inspection Agency (CFIA), Lethbridge, Canada;eUniversity of Homburg/Saar, Homburg, Germany; f German PrimateCenter, Goettingen, Germany

CONTACT Karla A. Schwenke [email protected] [email protected]

ABSTRACT

Although Chronic Wasting Disease (CWD) wasdetected in cervids in the late 1960s, the risk of CWDtransmission from cervids to humans is still puzzling.The zoonotic potential which might emanate fromCWD was studied by several research groups using amultitude of in vitro and in vivo approaches. Thesestudies provided evidence for a significant transmissionbarrier for CWD between cervids and humans, but donot allow the definite exclusion of such inter-speciestransmission. Old- world monkeys such as macaquesare currently considered the most reliable animal modelfor testing the zoonotic potential of CWD prions. Usingoral and parenteral transmission routes, our researchconsortium challenged cynomolgus monkeys withcharacterized CWD material starting 10 years ago. Totest the presence or absence of prion infection in thesemacaques, we injected bank voles intracerebrally withhomogenized brain, spinal cord and spleen tissue frommonkeys sacrificed 5–7years after CWD inoculation.Bank voles have been shown to be a universal acceptorfor prions derived from various species and were foundto be particularly susceptible to infection with CWDprions. Therefore, in our study bank vole bioassaysprovide a method of choice for the detection of prioninfection in the CWD-challenged cynomolgus mon-keys. Potential prion disease of macaques will bedetected by assessment of prion-specific clinical signsin bank voles. The clinical assessment includes weightmonitoring and routine observation of the animalsaccording to a clinical score sheet. Further, CNS andspleen tissue will be examined post mortem for PrPres

by Western blot analysis. In case of ambiguous results,we plan to perform second passage inoculations inbank voles. Taken together, these studies are expectedto contribute to a definite evaluation of the zoonoticrisk of CWD.

118. Effect of route of infection and PRNPgenotype on detection of preclinical infection usingblood samples from BSE-infected sheep

M. K. F. Salamata, O. Andreolettib, S. McCutcheona,A. R. A. Blancoc, J. C. Mansona, E. F. Houstona

aThe Roslin Institute, R(D)SVS, University of Edinburgh, Easter Bush,Midlothian, EH25 9RG, UK; bUMR INRA ENVT 1225, Ecole NationaleVétérinaire de Toulouse, Toulouse, France; cComponents DevelopmentLaboratory, NHS Blood and Transplant, Cambridge, UK

CONTACT E. F. Houston [email protected]

62 ABSTRACT

ABSTRACT

The possibility of on-going blood-borne transmissionof variant Creutzfeldt-Jakob disease (vCJD) is a majorconcern for public health authorities, particularly inlight of uncertainties surrounding the prevalence ofsubclinical vCJD infection in the human population.A robust and highly sensitive assay that can be appliedto readily accessible samples (e.g. blood, urine), fordetection of infected individuals before the onset ofdisease, would be of enormous benefit in preventingfurther transmission of vCJD. Using sheep experimen-tally infected with BSE as a model, we have previouslydemonstrated that the protein misfolding amplificationassay (PMCA) can detect seeding activity in bloodsamples as early as 6 months post-infection. However,we also observed significant differences in incubationperiod, and transmission of infection by transfusion,associated with genetic variation at PRNP codon 1411.The aim of this study was to determine the relationshipbetween codon 141 PRNP genotype and the onset ofdetection of seeding activity in preclinical blood sam-ples from orally-infected (donor) and intravenously-infected (recipient) sheep.

Samples were analysed using a microplate-based, min-iaturized bead serial PMCA protocol2−3, with brain homo-genate from tgShpXI transgenic mice expressing sheepARQ PrP (with added dextran sulphate) as substrate, andundiluted buffy coat samples from BSE-infected and unin-fected sheep as seed. In orally challenged sheep, bloodsamples were collected at intervals from 2 months post-infection until the onset of clinical signs. The first timepoint at which blood samples tested positive was veryvariable, ranging from 4 months to 12 months post-infec-tion and there was no obvious correlation between codon141 genotype and the onset of detection. In sheep infectedby blood transfusion, the first preclinical blood sampleswere collected at 6 months post-infection, and majority ofanimals tested gave positive PMCA results at this timepoint, regardless of codon 141 PRNP genotype. For bothorally and intravenously-infected sheep, all samples col-lected at time points following the onset of detection testedpositive in the PMCA. The results demonstrate that theroute of infection appears to have a much more significantinfluence than PRNP genotype on the onset of detection ofseeding activity in preclinical blood samples. They alsodemonstrate that once seeding activity is detected inblood samples from infected individuals, it is consistentlydetected in samples from subsequent time points through-out the incubation period. This data provides valuableinsights into the factors that may influence the sensitivityof detection of preclinical prion infection using blood-based assays.

References

[1] Tan, et al. J Gen Virol. 2012;93(Pt 12): 2749–2756[2] Moudjou, et al. MBio. 2013 31;5(1):e00829–13[3] Lacroux, et al. PLoS Pathog. 2014;10:e1004202.

119. Host genetic factors impacting prion diseases:genomic analysis of phenotypically distinct bovinespongiform encephalopathy cattle

Sandor Dudasa,b, Graham Plastowc, James Crossb andStefanie Czuba,b

aCanadian Food Inspection Agency, National Center for AnimalDisease Lethbridge; bUniversity of Calgary, Faculty of VeterinaryMedicine; cUniversity of Alberta, Agriculture, Food and NutritionalScience

CONTACT Sandor Dudas [email protected]

Populations and development of diagnostics and effec-tive therapeutics for AD.

ABSTRACT

Background/Introduction: Bovine spongiform ence-phalopathy (BSE)-challenged cattle have variable sus-ceptibility, incubation times and in rare cases, evendifferent disease phenotypes; our initial results suggestthis may be related to host genetic factors. Attemptshave been made to link bovine spongiform encephalo-pathy susceptibility with genotype using BSE field cases.These sample populations have a number of uncon-trolled variables making analysis difficult and likelycontribute to a lack of agreement between studies.Materials and Methods: To identify genetic factorsimpacting BSE in a more controlled population, tissuesamples from BSE-challenged cattle have been collectedfrom the APHA (UK), the FLI (Germany), the USDA(USA), and the CFIA (Canada). DNA from these tis-sues are currently being genotyped to identify regionsof the bovine genome which are associated with differ-ent BSE phenotypes. Once identified, targeted nextgeneration sequencing will be used to find functionallyrelevant genetic changes. If important polymorphismsare found, gene-edited cell culture will be used todemonstrate the impacts these genetic variants haveon prion expression and disease progression.Results: Genotyping of short incubation BSE challengecattle shows a significant link with the deletion genotypeat a previously uncharacterized prion gene promoterlocation. Functional consequences of this genotype arecurrently being determined using a promoter luciferaseassay. Non-prion genomic locations have also been iden-tified as unique in abnormal and/or atypical BSE cases.These include the previously identified sites BTA6-97.6Mb (ANTXR2/BMP3), BTA10-21.4Mb (CHMP4A/

PRION 63

MYH6), BTA20-4.90Mb (CART/STC2), and BTA20-39.85Mb (GDNF/SLC45A2) [1, 2]. We have also identi-fied additional non-prion genomic regions unique to theatypical/abnormal BSE cattle genotyped so far. Oneexample is a missense mutation at codon 439(G->S) inthe phosphoserine rich protein encoded by the kiaa0232gene. This amino acid (439G) is part of a conservedregion in species naturally affected by prion diseasesincluding sheep, goats and deer. The kiaa0232-encodedprotein has also been identified as a binding partner ofYHWAE/YHWAZ, members of the 14-3-3 proteinfamily involved in signal transduction and apoptosis [3].Conclusions: Genetic analysis of cattle with variableincubation or abnormal/atypical BSE could result inthe identification of proteins/pathways involved inprion disease pathogenesis. Introducing these function-ally relevant polymorphisms into cell culture modelswill hopefully result in changes in prion production/accumulation providing evidence for an altered diseaseprocess. This will further our understanding of pro-cesses important in BSE progression and could providefuture direction for selective breeding or therapeutics.

References

[1] Murdoch, et al. BMC Genet. 2010;11:20.[2] Thomson, et al. Prion. 2012;6(5):461–469.[3] Bandyopadhyay, et al. Nature methods. 2010;7

(10):801–805.

120. Detection of CWD prion seeding activity infaeces demonstrates both consistent shedding andpotential for environmental monitoring

Joanne M. Tennanta, Manci Lia, Davin M. Hendersona,Nicholas J. Haleyb, Candace K. Mathiasona andEdward A. Hoovera

aPrion Research Center, Department of Microbiology, Immunology,and Pathology, College of Veterinary Medicine and BiomedicalSciences, Colorado State University, Fort Collins, CO, USA;bDepartment of Microbiology and Pathology, Midwestern StateUniversity, Glendale, AZ, USA

CONTACT Joanne M. Tennant [email protected]

ABSTRACT

Background: Chronic wasting disease (CWD) is spread-ing in susceptible cervid populations in North America,Korea, and Europe. Environmental contamination andexposure to prions is considered to be a significant factorin horizontal CWD transmission. Currently disease sur-veillance is limited by the necessary presence and testabi-lity of infected animals. Previous studies have shown thatexcreta from CWD infected deer and elk contains prion

seeding activity. The detection of CWD through excreta,specifically faeces, could be beneficial in passive monitor-ing of CWD prevalence in endemic and emerginghabitats.Methods: Longitudinal collections of faeces fromlow-dose CWD inoculated deer were evaluated byreal-time quaking induced conversion (RT-QuIC).To emulate common environmental weathering con-ditions, CWD-positive faecal samples were driedand exposed to UV light and prion seeding activitywas evaluated by RT-QuIC before and after treat-ment. In addition, faeces from premises known tocontain either CWD positive or negative cervidswere collected and tested in RT-QuIC for prionseeding activity in a blinded experiment.Results: CWD prion seeding activity was detected by RT-QuIC in faeces of five out of six low-dose CWD inocu-lated 96GGdeer. The RT-QuIC detectable prion sheddingin these deer corresponded with detection of PrPCWD byIHC in rectoanal mucosa-associated lymphoid tissue(RAMALT) biopsy and prion shedding was detected dur-ing both the pre-symptomatic and symptomatic stages ofdisease. Prion seeding activity was little affected by dryingand exposure to ultraviolet light as both rectal collectedand dried faeces showed prion seeding activity. Faecescollected from the pens containing CWD positive vs.negative deer correlated with presence vs. absence prionseeding activity, respectively.Conclusion: These studies demonstrate that low-doseinoculated deer shed prion seeding activity in faecesthrough much of their disease course. Further, theprion seeding activity in faeces was still detectableafter exposure to UV light and desiccation. These find-ings suggest that environmental monitoring of CWDvia landscape faecal deposits may be possible as ameans for monitoring CWD in populations of free-ranging deer and elk.

121. Tau prion strain propagation and cytotoxicitydefined by live-cell imaging

Sang-Gyun Kanga,b, Ghazaleh Eskandari-Sedighia,c,Nathalie Daudea,b, Hristina Gapeshinaa, Jing Yanga

and David Westawaya,b,c

aCentre for Prions and Protein Folding Diseases; bDivision ofNeurology; cDepartment of Biochemistry, University of Alberta,Edmonton, Canada

CONTACT Sang-Gyun Kang [email protected]

ABSTRACT

Introduction: Tauopathies are a group of neurodegenera-tive disorders including Alzheimer’s disease (AD),

64 ABSTRACT

progressive supranuclear palsy (PSP), corticobasal degen-eration (CBD), and frontotemporal dementia (FTD).Tauopathies can be heterogeneous in clinical and patholo-gical presentation and may be characterized by the intra-cellular aggregation of microtubule-associated protein Tauin neurofibrillary tangles (NFTs). Also, abnormally phos-phorylated Tau becomes dissociated from neuronal micro-tubules and accumulates in paired helical filaments (PHFs).The conformational changes of monomeric soluble Tauinto oligomers and NFTs are thought to contribute toneuronal dysfunction and eventually cell death. As prion-like propagation of Tau (‘strain’ effects) has been invokedto explain phenotypic diversity in Tau pathogenesis, wehave assessed this possibility using a cell culture system.Materials and Methods: Previously, we reported dif-ferent pathologic variations (classes 1–5) in low-expres-ser TgTauP301L congenic mice encoding human Tau(2N4R, Isoform Tau-F) [1]. Sarkosyl-insoluble Taufractions (hereafter, referred to as Tau fibrils) wereprepared from mouse brains of each pathology class.HEK293 cells stably expressing a YFP-tagged humanTau 4R repeat domain (RD) fragment that includesaggregation-prone mutations (P301L/V337M, LM;proaggregation) (‘HEK-TauRD-LM-YFP cells’) [2]were transduced with liposome-protein complexesderived from each class of Tau fibrils. RecombinantTau RD fibrils (recTauRD) were also assessed.Transduced cells were split into six-well plates for themeasurement of insoluble Tau and for live-cell imaginganalysis.Results: Each class of Tau fibrils (including recTauRD)could show heterogeneous morphologies for Tau inclu-sions in the HEK293-TauRD-LM-YFP cells.Nonetheless, we observed three putative Tau prionstrains from brain samples as defined by striking differ-ences in their subcellular distribution of Tau deposits; alarge mass of aggregated Tau with no specific pattern(amorphous: ES1), juxtanuclear and nuclear membraneinclusions (juxtanuclear: ES2), and granular nuclearinclusions (speckles: ES3). Live-cell imaging analysisrevealed that ES1 and ES2 forms were toxic to thecells, whereas cells with ES3-type inclusions showedno cell death over a period of 16 h. Interestingly,nuclear blebs were prominent and co-localized withES1 and ES2 Tau fluorescent signals, indicating Tau-induced nuclear damage.Conclusions: TgTauP301L mice, an inbred congenicmodel of genetic Tauopathy, generate more than oneform of biologically active Tau, as revealed by differingpatterns of propagation and cytotoxicity in the cell-based assays. We also conclude that nuclear deforma-tion contributes to neurotoxicity in a sub-set ofTauopathies.

References

[1] Eskandari-Sedighi, Daude, et al. Mol Neurodegener. 4October 2017;12(1):72.

[2] Kaufman, Sanders, et al. Neuron. 23 November2016;92(4):796–812.

122. Diagnosis and biomarkers of Parkinson’s andDementia with Lewy bodies: rapid and ultra-sensitive quantitation of disease-associatedα-synuclein by αSyn RT-QuIC

Christina D. Orrùa, Bradley R Grovemana, Andrew GHughsona, Lynne D Raymonda, Gianluigi Zanussob,Bernardino Ghettic, Katrina J Campbella, Natália doCarmo Ferreiraa, Jiri Safard, Douglas Galaskoe, OskarHanssonf, g and Byron Caugheya

aLaboratory of Persistent Viral Diseases, Rocky MountainLaboratories, National Institute of Allergy and Infectious Diseases,National Institutes of Health, Hamilton, MT, USA; bDepartment ofNeurosciences, Biomedicine and Movement Sciences, University ofVerona, Verona, Italy; cIndiana University School of Medicine,Indianapolis, IN, USA; dDepartment of Pathology, Case WesternReserve University School of Medicine, Cleveland, OH, USA;eDepartment of Neurosciences, University of California-San Diego,La Jolla, CA, USA; fClinical Memory Research Unit, Department ofClinical Sciences Malmö, Lund University, Lund, Sweden; gMemoryClinic, Skåne University Hospital, Malmö, Sweden

CONTACT Christina D. Orrù [email protected]

ABSTRACT

Diagnosis of synucleinopathies such as Parkinson dis-ease and dementia with Lewy bodies can be aided byassays for the detection and quantification of patho-genic disease-associated forms of α-synuclein (αSynD).These assays need to be sufficiently sensitive, specific,and practical for analysis of accessible diagnostic sam-ples. Two recent αSynD seed amplification assays haveprovided valuable prototypes for detection of αSynD inpatients’ cerebrospinal fluid (CSF). However, theseassays require 5–13 days to perform. We have devel-oped an improved α-synuclein real time quaking-induced conversion (αSyn RT-QuIC) assay with similarsensitivity and specificity that provides quantitativeresults in 1–2 days. Our blinded analysis of cerebrosp-inal fluid from 29 synucleinopathy cases (12Parkinson’s and 17 dementia with Lewy bodies) and31 non-synucleinopathy controls, including 16Alzheimer’s cases, yielded 93% diagnostic sensitivityand 100% specificity. End-point dilution analysesdetected αSynD in as little as 0.2 μL of CSF.Furthermore, our test allowed us to quantitate relativeamounts of αSynD in CSF samples. We are currentlytesting additional well-characterized blinded panels ofCSF samples from individuals diagnosed with

PRION 65

synucleinopathies and controls. Moreover, we have alsodeveloped a capture protocol for the detection of αSynD

in human plasma spiked with αSyn positive brainhomogenate and in endogenous CSF samples fromindividuals affected by synucleinopathies. We can nowisolate αSynD from several biological samples for sensi-tive and specific detection by αSyn RT-QuIC.

123. Detection of prion seeds throughout the eyesof sporadic Creutzfeldt-Jakob disease patients

Christina Orrua, Katrin Soldaub, Christian Cordanoc,Jorge Llibre-Guerrad, Ari J. Greene, Henry Sanchezc,Bradley R. Grovemana, Steven D. Edlandf,g, Jiri G.Safarh,i, Jonathan H. Linb, Byron Caugheya, MichaelD. Geschwindc and Christina J. Sigurdsonb,j

aLaboratory of Persistent Viral Diseases, Rocky Mountain Laboratories,National Institute of Allergy and Infectious Diseases (NIAID), NationalInstitutes of Health (NIH), Hamilton,MT, USA; bDepartment of Pathology,University of California, San Diego, La Jolla, CA USA; cDepartment ofNeurology, Memory and Aging Center, University of California, SanFrancisco (UCSF), San Francisco, CA, USA; dCognitive and BehavioralResearch Unit, National Institute of Neurology, Havana, Cuba;eDepartment of Neurology, Multiple Sclerosis Center, University ofCalifornia, San Francisco (UCSF), San Francisco, CA, USA; fDepartmentsof Family Medicine & Public Health and gNeurosciences, University ofCalifornia, San Diego, La Jolla, CA USA; hDepartments of Pathology andiNeurology, Case Western Reserve University, Cleveland, OH, USA;jDepartment of Pathology, Immunology, and Microbiology, Universityof California, Davis, Davis, CA, USA

CONTACT Christina Orru [email protected]

Orrù and Soldau, mBIO 2018 9:e02095–18. https://doi.org/10.1128/ mBio.02095-18

ABSTRACT

Cases of iatrogenic Creutzfeldt-Jakob disease (CJD) havebeen reported from corneal transplants, yet the levels ofprions in the eye remain unknown. Approximately, 40%of sporadic CJD patients develop visual symptoms andoften seek ophthalmological consultation. This studyprobed the occurrence, level, and distribution of prionsin the eyes of patients with sCJD, the most commonprion disease in humans. We used the highly sensitivereal time quaking-induced conversion (RT-QuIC) assayto measure post-mortem levels of prion seeding activityin cornea, lens, ocular fluid, retina, choroid, sclera, opticnerve, and extraocular muscle. This is the largest seriesof sCJD patient eyes studied by any assay to date. Wedetected prion seeding activity in 100% of sCJD eyesrepresenting three common sCJD subtypes with a fourlog-fold range of seeding activity observed across indi-viduals. The retina consistently showed the highest prion

seeding activity. Within the retina, prion deposits werealso detected by immunohistochemistry in the outerplexiform layer, and in some eyes the inner plexiformlayer. With increased distance from the brain prion seedlevels by RT-QuIC declined in eye tissues. Collectively,these results reveal that sCJD patients accumulate prionsthroughout the eyes, indicating the potential diagnosticutility as well as the possible biohazard.

124. Longitudinal studies of CWD after low-doseoral exposure in white-tailed deer

Clare E. Hoover*, Nathaniel D. Denkers, Kristen A.Davenport, Davin M. Henderson, Amy V. Nalls, ErinMcNulty, Joanne Tennant, Sarah Cooper, Manci Li,Lauren Bracchi, Candace K. Mathiason and EdwardA. Hoover

Prion Research Center, Department of Microbiology, Immunology,and Pathology, College of Veterinary Medicine and BiomedicalSciences, Colorado State University, Fort Collins, CO, USA

CONTACT Edward A. Hoover [email protected]

*Present address: AstraZeneca, Waltham, NJ

ABSTRACT

Background: The facile transmission of chronic wast-ing disease (CWD) in North America, Asia and Europecontinues despite exposure to very low concentrationsof prions shed by infected cervids in secreta andexcreta. Explanations for this enigma could be thatexcreted prions may have enhanced infectivity and/orthat the infectious prion dose is just quite low.Historical studies exploring CWD pathogenesis indeer have used exposures to CWD-positive brainhomogenates containing from 0.5 to 10 g of brain –doses much higher than those measured in excreta, andthereby likely to ever occur in nature. We therebysought to more closely emulate natural exposure con-text in our current experimental studies.Methods: To explore how the origin or infectious dose ofCWD prions may influence disease transmission orpathogenesis, we orally exposed cohorts of n = 4/groupwhite-tailed deer to low doses of CWDpresented as eitherCWD-positive brain homogenate (containing either 1 mgor 300 ng of brain) or to an amount of pooled saliva fromCWD+ donors and containing prion seeding activity (byRT-QuIC) equivalent to 300 ng of CWD+ brain [1].Inoculated deer were then longitudinally monitored for:(a) onset of prion infection in biopsied tonsil and recto-anal lymphoid tissues by RT-QuIC and immunohisto-chemistry; (b) prion shedding in saliva and faeces (by

66 ABSTRACT

RT-QuIC); (c) onset of clinical signs (weekly observationand scoring); and (d) time to clinical disease.Results: We first detected CWD infection by RT-QuICseeding activity in tonsil biopsies at 6 or 9 months postexposure, in all inoculation cohorts. Prion shedding insaliva was detected (by RT-QuIC or PMCA-RT-QuIC[2]) concurrent with the first positive tonsil biopsy.Detection of prion seeding activity in faeces was lesscommon overall and its onset observed after positivityin recto-anal lymphoid tissue biopsies. In comparisonto historical studies, in which >1,000-fold higher inocu-lation doses were used, the time to first detection oninfection (by tonsil biopsy) was prolonged by~6 months. However, among the low-dose inoculationcohorts in this study, neither the attack rate, prionshedding profile, nor long-term disease course differedsignificantly, including the deer exposed to prions ofsaliva vs. brain origin.Conclusions: These studies demonstrate that (a)much lower doses of CWD prions that have beenused historically for point source exposure studiesare sufficient to induce prion infection, shedding,and disease; and (b) although time to first detect-able positivity is lengthened, the qualitative aspectsof disease pathogenesis thereafter are indistinguish-able by inoculum source or exposure magnitude.

Our future studies will aim at establishing the mini-mum oral infectious dose for CWD in deer and onexploring exposure cofactors.

References

[1] Hoover CE, et al. in preparation, 2019[2] Davenport KA, et al. J Clin Microbiol. 27 August

2018;56(9).Supported by NIH R01-NS-061902, P01-AI-077774,F30-ODO-118,143, T32-OD0-10,437

125. Bioassay confirms the relevance of surf-PMCAfor human prion decontamination studies

Maxime Bélondradea, Simon Nicota, Christelle Jasa,b,Vincent Béringueb, Chantal Fournier-Wirtha, SylvainLehmannc and Daisy Bougarda*aPathogenesis and control of chronic infections, EtablissementFrançais du Sang, Inserm, Université de Montpellier, Montpellier,France; bInstitut National de la Recherche Agronomique (INRA),UR892, Virologie Immunologie Moléculaires, Jouy-en-Josas,France; cCHRU de Montpellier and Université de Montpellier,IRMB, INSERM U1183, Laboratoire de Biochimie ProtéomiqueClinique, Montpellier, France

CONTACT Daisy Bougard [email protected]

Abstract not available.

126. Dickinson, the forgotten man of the prionstory

James Hopea and Jason Bartzb

aOne Health, Norwich, Norfolk, UK; bCreighton University, Omaha,Nebraska, USA

CONTACT James Hope [email protected]

Alan Dickinson, geneticist, born 30 March 1930; died27 September 2017

Alan Dickinson died just after the Prion 2017meeting in Edinburgh [1] and, sadly, his iconoclasticstance (on everything) and working tenet of theimportance of being awkward, has meant his contri-bution to our prion world has faded much tooquickly. To redress the balance a wee bit, let’s beclear: he was a fantastic geneticist and, with HughFraser and Moira Bruce, sorted out much of theconfusion and mystery of scrapie in sheep thatraged in the late 1950s and 1960s. His post-graduatetraining in genetics was meticulously applied to thebreeding of mice and sheep, and he was able tobiologically define alleles of a genetic locus (Sinc inmice) in both which appear to control the timing andtempo of the inoculated (and natural) disease withclockwork precision. The methods of strain typingusing vacuolar pathology and mice of defined prionprotein genotype (PrP is a product of the Sinc locus)are still used today in prion and prion-like diseaseresearch, and strain theory in TSEs and other neuro-degenerative disease continues to intrigue and testour understanding of mechanisms of propagation,blocking and phenotype stability. It is interesting tospeculate how much of his ‘awkward’ legacy has beenexplained by prions. What is our molecular explana-tion for breakdown of the 87A strain to ME7? Howdoes one slow replicating strain prevent disease withthe characteristics of a later-inoculated, short-incuba-tion period prion? Current work does indicate thatprion strains compete for PrPC in a common

PRION 67

population of cells [2], consistent with AGD’s origi-nal ‘replication site’ hypothesis [3]. And ‘strain-spe-cific’ co-factors, the prion’s host-independentgenome, are still sought after today, but remain elu-sive [4]. Crucially, he did use his insights for prac-tical good, inter alia predicting the dangers of humangrowth hormone (hGH) therapy for pituitary dwarf-ism, and validating methods for its safe purification[5], decades before the current controversy of hGHtherapy, ‘Alzheimer’ prions and Aβ pathology. Hedied continuing to search for an elusive second mole-cule to explain these biological phenomena – theiconoclast in him never accepted PrP (or the nomen-clature) as the sole component of a naturally-infec-tious prion – and he ruffled a few feathers in old ageby continually defining molecular biology as ‘fishingwithout a licence’, rephrasing Chargaff’s view of it as‘the practice of biochemistry without a licence’ [6].Even so, his life’s work deserves recognition to thisday in the bibliographies of our publications.

KEYWORDS: virino; strains; Sinc; mouse; genetics;vacuolation; stability classes; hGH; history of science

References

[1] Green, E. Guardian (Manchester, UK) 23 November2017. Geneticist who carried out groundbreakingresearch into the behaviour of diseases including scra-pie and CJD [cited 15 February 2018].

[2] Eckland TE, Shikiya RA, Bartz JC. Independent ampli-fication of co-infected long incubation period low con-version efficiency prion strains. PLoS Pathog. 2018;14:e1007323.

[3] Dickinson, AG, Outram GW. The scrapie replication-site hypothesis and its implications for pathogenesis.In: Prusiner SB, Hadlow WJ, editor, Slow transmissiblediseases of the central nervous system. Vol. 2. NewYork, NY: Academic Press, Inc.; 1979. p. 13–31.

[4] Saá P, Sferrazza GF, Ottenberg G, et al. Strain-specificrole of RNAs in prion replication. J Virol.2012;86:10494–10,504.

[5] Taylor DM, et al. Preparation of growth hormone freefrom contamination with unconventional slow viruses.Lancet. 1985 Aug 3; 8449:260–262.

[6] Wright, P. Guardian (Manchester, UK) 2 July 2002.Disillusioned biochemist who pioneered our under-standing of DNA [cited 7 February 2019].

127. Focal and diverse deposition in a mousemodel of 4-repeat tauopathy is paralleled bydistinct biochemical and cellular signatures; closerelationship to Tau species in P301L patients

D. Westaway, N. Daude, G. Eskandari-Sedighi, S. Kang,C. Kim, S. Fleck, J. Yang, H. Wille, E. Gelpi and J. G Safar

ABSTRACT

Background: Mutations in the Tau (MAPT) gene cancause neurodegenerative diseases such as frontotemporallobar degeneration (FTLD) but, strikingly, patients withthe same mutation have very different clinical phenotypes;the cause of this diversity is not understood.Heterogeneities have also been observed in a transgenic(Tg) mouse line expressing low levels of mutant humanP301L Tau and backcrosses of founder stocks of mice toinbred genetic backgrounds to attenuate contributionsfrom modifier loci failed to eliminate variability in dis-ease-related phenotypes (Eskandari-Sedighi et al., 2017).Here biochemical and cellular assays have been used toassess chemical and biological characteristics of P301L Tauspecies from Tg.P301L mice and from human FTLD.Methods: We used Tg.P301L mice from C57BL/6Tac,129/SvEvTac and FVB/NJ genetic backgrounds andbrain material from eight Iberian FTLD patients withthe same P301L mutation and similar MAPT locushaplotypes (Borrego-Ecija et al., 2017). Tau specieswere profiled using microfluidic western assays, immu-nostaining, conformation-dependent immunoassays(CDI), conformational stability assays (CSAs), trypsindigestion, as well as by kinetic and morphological per-formance measures in a cell-based seeding assay.Electron microscopy (EM) was used to investigate fibrilmorphology.Results: TgP301L mice yielded six patterns of Taudeposition in neurons and astrocytes; these includedbrain stem and focal deposition and in Class VI,present in half of aged mice, tangle-like deposits inthe locus coeruleus. As defined by biochemical assays,three distinct Tau signatures were present in thebrains of inbred Tg mice, these signatures often cor-relating with pathology classes. EM revealed thatbrains of Classes I, II, III and IV animals exhibit atleast three distinguishable fibril morphology; themost abundant being straight filaments with twistedribbon-like filament and coiled filaments being pre-sent in some classes. Analyses of detergent-insolubletau in the frontal cortex of FTLD patients demon-strated remarkable conformational heterogeneity withat least two Tau signatures that mimicked a CSApattern found in a subset of aged TgP301L mice.Toxicity above baseline in transfected cells was great-est with brain-derived material, with different pre-parations differing in their kinetics of cell loss andfluorescent signals at the nuclear margin.Conclusions: Heterogeneities in tau pathology and che-mical signatures were observed within congenic stocksof TgP301L mice and in different P301L patients; cru-cially, signatures present in patients also manifested in

68 ABSTRACT

a subset of mice. We conclude that our Tg modelreveals non-synchronous, stochastic chemical eventsthat initiate tau deposition and that different toxicityprofiles of Tau isolates affect their ability to propagate.

128. Strain profiles of CJD in venison consumersand non-consumers from Alberta andSaskatchewan

Jennifer Myskiwa,b, Lise Lamourieuxa, MichaelCoulthartc, Valerie Simd and Stephanie Bootha,b

aZoonotic Diseases and Special Pathogens, National MicrobiologyLaboratory, Public Health Agency of Canada, Winnipeg;bDepartment of Medical Microbiology and Infectious Diseases,University of Manitoba, Winnipeg; cCanadian CJD SurveillanceSystem, Public Health Agency of Canada, Ottawa; dDivision ofNeurology, Department of Medicine Centre for Prions and ProteinFolding Diseases, University of Alberta, Edmonton

CONTACT Jennifer Myskiw [email protected]

ABSTRACT

Chronic wasting disease (CWD) is a fatal neurodegen-erative disease that is spreading rapidly through wildcervid populations in Alberta and Saskatchewan. Theepidemic of CWD in Canada not only has implicationsfor tourism and hunting but it is also a concern overpossible zoonotic transmission to humans who eatvenison from infected deer. When bovine spongiformencephalopathy (BSE) reached epidemic proportions,variant CJD was identified as an acquired form ofBSE due to the unique biochemical fingerprint of thepathologic prion protein (PrP). While the CanadianCJD Surveillance System (CJDSS) is staying vigilant toidentify CWD human cases, it is impossible to knowwhat the phenotype of human CWD prions wouldpresent with or whether they would be distinct fromsporadic CJD cases. Additionally, there is currently noconvenient laboratory model to study these differencesexperimentally. For these reasons we are undertaking asystematic analysis of the molecular diversity of CJDcases in patients who resided in Alberta andSaskatchewan at their time of death. We are comparingCJD profiles of venison consumers and non-consumersusing a variety of clinical imaging, pathological andbiochemical markers. Firstly, MRIs have been reviewedand neuropathology of over 40 patients for whom CJDwas confirmed as the cause of death analysed. In thisway we are able to select clinically affected areas forfurther biochemical analysis of prion protein. We willpresent data including our analysis to date of levels ofprotease sensitive and resistant prion protein, tempera-ture and detergent denaturation profiles, and glycoformtyping. By combining these methodologies, we intend

to extend the baseline CJD typing that is already com-pleted by the CJDSS. Importantly we aim to increaseour understanding of the human prion strains thataffect the Canadian population. By doing so, we willestablish a baseline for the identification of any futureatypical CJD cases. For example, those that could ariseas a result of exposure to CWD.

129. Deregulation of SINE RNA levels inAlzheimer’s disease patients

Yubo Chenga, Luke Savillea, Babita Gollena, MitchellLiamb, Christopher Isaaca, Jogender Mehlab,Mohajerani Majidb and Athanasios Zovoilisa,b

aDepartment of Chemistry and Biochemistry, University ofLethbridge, Lethbridge, Canada; bDepartment of Neuroscience,University of Lethbridge, Lethbridge, Canada

CONTACT Athanasios Zovoilis [email protected]

ABSTRACT

As the human life span increases, the number of peoplein Canada suffering from Alzheimer’s disease (AD) isexpected to rise dramatically. A number of studies haverevealed that gene expression networks in AD arederegulated. However, the molecular mechanisms asso-ciated with this deregulation remain largely unknown. Ina previous work, we have shown that mechanisms invol-ving non protein-coding RNAs, such as ShortInterspersed Nuclear Element (SINE) RNAs (Zovoiliset al., Cell 2016) are key in maintaining transcriptionalhomeostasis in cells. Recently, we showed that in mouse,RNA from one of the most frequent SINE repeat classes(B2 RNA) controls expression of immediate early genes(IEGs) through binding and inhibition of RNAPolymerase II. However, it remained unclear whetherderegulation of this mechanism could underlie a patho-logical condition in human. Here, we applied an inte-grative RNA genomics and bioinformatics approach todissect any connection between SINE RNAs andAlzheimer’s disease. Our study revealed that SINERNAs are subject to abnormal processing in humanAD patients and are connected with deregulation ofgene expression.

130. Evaluation of the sensitivity of different RT-QuIC substrates in detecting and characterizingCWD prions in brains of Norwegian cervids

Edoardo Bistaffaa, Tram Thu Vuongb, Linh Tranb,Federico Angelo Cazzanigaa, Giulia Salzanoc,Giuseppe Legnamec, Giorgio Giacconea, SylvieBenestadb and Fabio Modaa

PRION 69

aDivision of Neurology 5 and Neuropathology, Fondazione IRCCSIstituto Neurologico Carlo Besta, Milano, Italy; bNorwegianVeterinary Institute, Oslo, Norway; cLaboratory of Prion Biology,Department of Neuroscience, Scuola Internazionale Superiore diStudi Avanzati (SISSA), Trieste, Italy

CONTACT Tram Thu Vuong [email protected]

ABSTRACT

Chronic wasting disease (CWD) is a highly contagiousprion pathology affecting captive and free-ranging cervidpopulations. From its first description 50 years ago, CWDhas been detected in United States, Canada, South Koreaand, most recently, in Norway. At present, we do not knowif these diseases are caused by the same prion strain orwhether they spontaneously arose in different countries.CWD spreading among cervids is one of the most impor-tant issue for public health due to the ability of this agent toinfect a large number of animals and the possible transmis-sion to other animal species, including humans and ovine.Indeed, possible transmission of CWD to ovine that share,in some countries, the same geographic areas of CWDinfected animals cannot be excluded. For this reason, it isof fundamental importance to develop a strategy allowingidentification of CWD infected samples and verify theirability to propagate within the same or different species. Tothis aim, we are optimizing RT-QuIC assay coupled withspecific biochemical analysis for evaluating whether differ-ent CWD samples could produce final reactions productswith peculiar differences useful for CWD strain’s identifi-cation. RT-QuICwere performed using different substratesof reaction (truncated PrP protein from hamster, reindeerand deer species) for evaluating their sensitivity and speci-ficity in detecting CWD in brains of Norwegianmoose, reddeer and reindeer. Moreover, through biochemical assess-ments (PK digestion, guanidine hydrochloride evaluationand use of antibodies against different PrP epitopes), weevaluated whether and to what extent final RT-QuIC pro-ducts were able to acquire distinct biochemical featuresaccording to the strain of CWD. Preliminaryresults showedthat deer PrP detected CWD in all species with higherefficiency compared to hamster and reindeer. Notably,truncated deer-PrP was able to rapidly detect positivesamples (within 3 h from the beginning of the reaction)compared to the other substrates. Although with less sen-sitivity, reindeer-PrP identified red deer, reindeer andmoose CWD, with this latter being characterized bylower resistance to PK digestion and different PrP bandingprofile (at Western blot) compared to the others.Therefore, RT-QuIC can be exploited for detecting CWDinfected species with high sensitivity and specificity, whilebiochemical data seem to indicate the possibility of identi-fying the species of CWD origin. If confirmed, such infor-mation will be of fundamental importance for planning

actions to contain the spread of the infection (especially toother species, like ovines) in CWD affected regions.

KEYWORDS: Prion; CWD; RT-QuIC

131. Targeting a regulatory subunit of proteinphosphatase 1 by Sephin-1 reduces prion infectionin neuronal cells

Simrika Thapa, Dalia. H. Abdelaziz, BasantAbdulrahman and Hermann M. Schatzl

Department of Comparative Biology & Experimental Medicine,Hotchkiss Brain Institute and Calgary Prion Research Unit, Universityof Calgary, Calgary, Alberta, Canada

CONTACT Simrika Thapa [email protected]

ABSTRACT

Prion diseases are infectious and fatal neurodegenera-tive diseases in human and animals caused by misfold-ing of the cellular prion protein (PrPC) into theinfectious isoform PrPSc. These diseases have the poten-tial to transmit within or between species, and no cureis available to date. Targeting the unfolded proteinresponse (UPR) as an anti-prion therapeutic approachhas been widely reported for prion diseases. Here, wedescribe the anti-prion effect of Sephin-1 which hasbeen shown to protect in mouse models of proteinmisfolding diseases including amyotrophic lateralsclerosis by selectively inhibiting the stress-inducedregulatory subunit of protein phosphatase 1, thusprolonging the eIF2a phosphorylation. In this study,we describe that Sephin-1 significantly reduced PrPSc

in persistently prion-infected neuronal cells, 22L-ScN2a, RML-ScN2a and RML-ScCAD5 cells.Moreover, prion seeding activity was reduced in treatedcells in RT-QuIC assay. No effect of Sephin-1 on thelevels of PrPC expression was observed in uninfectedN2a cells. Importantly, we found that Sephin-1 signifi-cantly overcame the ER stress induced in treated cellsas measured by lower expression levels of stress-induced aberrant PrP and stress markers (BiP andCHOP). At present, we are investigating effectivenessof intraperitoneal Sephin-1 treatment against RMLprions in vivo. These results suggest that Sephin-1could be a potential anti-prion drug selectively target-ing one component of UPR pathway.

132. Metformin ameliorates prion infection inneuronal cells by enhancing autophagy

Dalia Abdelaziz, Simrika Thapa, Basant Abdulrahmanand Hermann Schatzl

70 ABSTRACT

Department Comparative Biology & Experimental Medicine, Facultyof Veterinary Medicine, Calgary Prion Research Unit, and HotchkissBrain Institute, University of Calgary, Calgary, Alberta, Canada.

CONTACT Dalia Abdelaziz [email protected]

ABSTRACT

Metformin is the first-line medication for the treatmentof type 2 diabetes and a well-known AMPK activator.Recently, substantial efforts have been invested inestablishing the role of metformin in the treatment ofneurodegenerative diseases, such as Alzheimer’s,amnestic mild cognitive impairment, Parkinson’s andHuntington’s disease. Metformin was shown to be neu-roprotective and to promote adult neurogenesis underboth physiological and pathological conditions in vivo.This prompted us to investigate the anti-prion effec-tiveness of metformin in two different persistentlyprion infected neuronal cell lines (RML-CAD5 and22L-N2a). We found that metformin treatment, atnon-toxic concentrations of 0.5 and 1 mM, decreasedthe PrPSc load in both 22L-N2a and RML-CAD5 cells,as shown by lower signals of PK resistant PrP inimmunoblots and less conversion activity in RT-QuIC. On the other hand, metformin treatment hadno effect on PrPC levels in uninfected N2a cells. Sincethere is a solid body of evidence showing autophagyenhancing activity of metformin, we decided to test thelevel of autophagy markers in the treated cells. Notably,the LC3 II signal was found to be upregulated in bothCAD5 and N2a cells after treatment with metformin,which confirms the autophagy inducing effect of met-formin in both neuronal cell lines. Currently, we areinvestigating the effect of intraperitoneal metformintreatment on the survival of RML-infected mice. Inconclusion, our findings describe for the first time theanti-prion effect of metformin, which can be at leastpartially explained by its autophagy inducing activity.In future studies, we are planning to do synergisticcombination of metformin with other anti-prion can-didate drugs to test if this will improve the anti-prionefficacy via working on different pathways.

133. Neurofilaments in blood: a new promisingbiomarker of scrapie

Marco Rossia, Elena Bozzettaa, Cristina Casalonea,Cristiano Coronaa, Maria C. Cavarrettaa, HenrikZetterbergb, Kaj Blennowb and Daniela Melonia

aIstituto zooprofilattico del Piemonte Liguria e Valle d’Aosta, Turin,Italy; bInstitute of neuroscience and physiology, University ofGothenburg, Gothenburg, Sweden

CONTACT Marco Rossi [email protected]

ABSTRACT

Introduction: In vivo accurate, easy-to-perform, andearly diagnostic tests are crucial for the identificationof prion subjects during their early or preclinicalphase of disease and finally for managing publichealth issues. Excellent biomarkers for prion diseaseshave been identified in the cerebrospinal fluid (CSF),but this tissue is difficult to get from sheep and istherefore of little utility for the control of scrapie.Recently, the measurement of neurofilaments lightchain (NF-L), a surrogate biomarker of neuroaxonaldegeneration, by the SIMOA ultra-sensitive singlemolecule array technology, revealed high NF-L levelsin blood of patients with preclinical stages of humanprion diseases. In this work, we adapted the SIMOAtest for the measurement of NF-L in blood of scrapieinfected sheep and healthy animals and it showed apromising utility of blood serum NF-L as biomarkerof scrapie in sheep.Materials and Methods: A set of 20 sheep blood serumblinded samples were tested by the SIMOA technology.Nine samples were from scrapie affected sheep and 11from healthy animals. The gold standard was assessedby testing the medulla oblongata of sheep both with anEU approved Elisa rapid test followed by a confirma-tory Western blot assay.Results: Results revealed a clear capacity of Simoa NFLtest to classify the two groups of samples according tothe presence or absence of scrapie infection. All thepositive samples according to the gold standard showedhigh NF-L levels, while only one of 10 samples fromhealthy animals showed NF-L values overlapping withpositive samples.Conclusion: These data suggest that neurofilamentsare sensitive and specific blood biomarkers for thediagnosis of scrapie in sheep and might representpromising tools for an early diagnosis. We are nowworking for establishing the basal levels of NF-L in alarger group of healthy sheep and in sheep with dif-ferent types of diseases other than scrapie. We are alsoworking for determining the variation of NF-L levelsin experimentally scrapie-infected sheep at variousintervals of the incubation period and at the terminalstages of disease.

134. The transmissible spongiformencephalopathies surveillance in small ruminantsin Romania for a period of 10 years

Florica Bărbuceanua, Ioana Neghirlab, TheodoraChesnoiub, Cristina Diaconua, Stefania-FeliciaBarbuceanuc and G. Predoid

PRION 71

aInstitute for Diagnosis and Animal Health Bucharest, București,Romania; bNational Sanitary Veterinary and Food Safety Authority,București, Romania; cFaculty of Pharmacy, Carol Davila University ofMedicine and Pharmacy, București, Romania; dFaculty of VeterinaryMedicine Bucharest, București, Romania

CONTACT Florica Bărbuceanu [email protected]

ABSTRACT

The scarpie belongs to the TSE group and is a fataldegenerative disease, affecting the central nervoussystem of ovine and caprine. There are two types ofscrapie: classical and atypical. The classical scrapieaffects the animals aged from 2 to 5 years and is verycontageous, while the atypical scrapie affects the ani-mals older than 5 years and is considered to have alow degree of infectivity. The aim of this study is topresent information on the Romanian TSESurveillance Program, performed in compliancewith the EU Commission and OIE requirements, aswell as the TSE test results in small ruminants (ani-mals of the category clinical suspicions, dead animalsand animals slaughtered normally) performed in thenational net for transmissible spongiform encephalo-pathies, between 2007 and 2017. Between 2007 and2017 were tested in Romania approximatively339,643 small ruminants. All the 787 cases of scrapiewere confirmed by the TSE NRL (National ReferenceLaboratory) by immunoblotting and immunohisto-chemical tests, with the performance of the discrimi-natory and genotyping testing. All animals diagnosedwith scrapie were affected by the classical form. Forall suspicion cases, a differential diagnosis was per-formed against rabies, Aujeszky disease andlisteriosis.

KEYWORDS: Classical scrapie; Romanian TSE surveil-lance program; NRL-TSE

135. Characterization of serum miRNA in prioninfected elk and hamsters reveal putativebiomarkers

Jessy Slotaa, Sarah Medinab, Megan Klassenc andStephanie Boothb

aUniversity of Manitoba, Winnipeg, Canada; bPublic Health Agency ofCanada, Ottawa, Canada; cPublic Health Agency of Canada – NML

CONTACT Jessy Slota [email protected]

ABSTRACT

Prion diseases are a group of fatal neurodegenerativediseases that arise in both humans and animals followingmisfolding of the cellular prion protein into a disease

associated structural conformation. Afflicted individualsexperience a decline inmental capacity owing to the deathof brain cells and spongiform change, although the phe-notypic characteristics of these diseases vary widely. Thisheterogeneity as well as a lack of robust diagnostic meth-odologies make the diagnosis of prion diseases very chal-lenging. It is especially important to develop betterdiagnostic assays for prion diseases as diagnosis mustprecede the onset of clinical signs and symptoms as wellas permanent neuronal damage for any potential thera-peutics to be of benefit. Micro-RNAs (miRNAs) are smallnon-coding RNA molecules which regulate expression ofmRNA through the RNA interference pathway and maybe found within cells as well as circulating biological fluidssuch as serum and cerebrospinal fluid (CSF).Identification of signatures of altered levels of specificmiRNAs within biofluids during disease states is a pro-mising new avenue of research which may fuel the devel-opment of improved diagnostic assays. Therefore,identifying patterns of miRNAs deregulated within bio-fluids such as serum during prion infection may lead tothe discovery of novel biomarkers, aiding in the diagnosisof these diseases. In the present study, Illumina next-generation sequencing was used to characterize themiRNA content of serum taken from Elk with chronicwasting disease (CWD) as well as hamsters infected with263K scrapie prions. This analysis was also applied to CSFtaken from the 263K infected hamsters. Multiple miRNAswere detected to be altered between both serum and CSFisolated from the infected animals compared to thehealthy control animals. Subsequent analysis revealed apattern of five miRNAs which were consistently down-regulated in serum samples taken from both elk andhamsters infected with prions, and therefore are strongcandidate biomarkers for prion diseases. These resultsmay lay the groundwork towards utilizing alteredmiRNA signatures as the basis for improved prion diag-nostic assays.

136. Identification of novel microRNA candidatesmediating alpha-synuclein uptake in cells

Elena De Ceccoa, Remo Sangesb, Luca Bragac, PaolaZagoa, Joanna Narkiewicza, Fabio Perissinottod, IreneGuadagninoe, Sandro Banfie, Mauro Giaccac andGiuseppe Legnamea,d

aLaboratory of Prion Biology, Department of Neuroscience, ScuolaInternazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy;bComputational Genomics Laboratory, Department ofNeuroscience, Scuola Internazionale Superiore di Studi Avanzati(SISSA), Trieste, Italy; cICGEB International Centre for GeneticEngineering and Biotechnology, Padriciano, Trieste, Italy; dELETTRA

72 ABSTRACT

Sincrotrone Trieste S.C.p.A, Basovizza, Trieste, Italy; eTIGEM TelethonInstitute of Genetics and Medicine, Pozzuoli, Napoli, Italy.

CONTACT Elena De Cecco [email protected]

ABSTRACT

Background: Alpha-synucleinopathies are a group offatal neurodegenerative disorders for which neithereffective therapies nor preclinical biomarkers are yetavailable. Indeed, one major characteristic of these syn-dromes is that the first clinical symptoms start to appearonly when more than 70% of dopaminergic neurons inthe substantia nigra and striatum have been lost, andbrain functions are irreversibly compromised. Hence,the identification of early-stage biomarkers poses as acrucial goal in the fight against the progression of neu-rodegeneration, as well as for the development of moreeffective and specific drugs. Dysregulation of the expres-sion of non-coding RNAs (ncRNAs) is a common fea-ture among several neurodegenerative diseases. MicroRNAs (miRNAs) are the most studied among ncRNAsand have been shown to participate in Multiple SystemAtrophy and Parkinson’s Disease pathogenesis at var-ious levels. This work aims at identifying miRNAs thatplay a role in the internalization of extracellular alpha-synuclein aggregates, which is a key step in the propaga-tion of synucleinopathies.Materials and methods: Our analysis focuses on theinternalization of short fibrillar species of alpha-synu-clein, as recent findings suggest that short fibres areeasily internalized by cells and mediate the spreading ofthe pathology. To this aim, we set up a comprehensivemiRNA screening approach that takes advantage ofhigh throughput screening technology and allows usto screen thousands of miRNAs at the same time. Theevaluation of the effect of each miRNA is based on thedifferential fluorescent labelling of internalized andnon-internalized aggregates.Results: Starting from a library of 2042 miRNAs, wefound several candidates, which, upon overexpression,either promote or inhibit the internalization of alpha-synuclein short fibrils. Top hits from each phenotypewere subjected to a detailed bioinformatics analysisresulting in the identification of multiple possible targetgenes, many of which are common targets of more tophit miRNAs. We are currently validating the role ofsome of the selected target genes in order to unravel thecontribution of new molecular pathways to the com-plex phenomenon of aggregates propagation.Conclusions: Our approach led to the identification of anumber of miRNAs involved in alpha-synuclein short

fibrils uptake, as well as of some target genes. Thesefindings represent a significant advancement to betterunderstand the complex miRNA regulation in diseasedbrains, and will eventually provide a starting point todesignmiRNA-based therapies against synucleinopathies.

137. Stem cells treatment ameliorate clinical signsand increase incubation periods in prion infectedrodents

Paulina Soto, Enrique Antonio Armijo Fuentes, CarlosKramm, Claudio Soto and Rodrigo Morales

Department of Neurology, The University of Texas Health ScienceCenter at Houston, Houston, TX, USA.

CONTACT Paulina Soto [email protected]

ABSTRACT

Prion diseases or transmissible spongiform encepha-lopathies are a family of rare progressive neurode-generative disorders that affect both humans andanimals. Currently, these disorders have no cure. Inthe last decades, great efforts have been made toestablish effective disease-modifying options to treatthem. In recent years, the potential use of inducepluripotent stem cells (iPS) derived neuroprecursors(NPs) has been suggested as a promising cell-replace-ment therapy for neurodegenerative disorders. iPS-derived NPs have been shown to be effective inanimal models of several neurodegenerative diseasesincluding Parkinson’s and Huntington’s diseases,multiple sclerosis, and amyotrophic lateral sclerosis.We tested the therapeutic potential of NPs on prioninfected rodents using different means of delivery.Briefly, C57BL/6J female mice were intra-craniallyinoculated with RML prions and the potential bene-fits of NPs was evaluated in three different formats:(1) intra-cerebral administration when the animalsshowed the first prion-associated clinical signs, (2)intra-venous treatment before the onset of symptoms,and (3) i.v infusions on early symptomatic subjects.Our results show that in all cases NPs significantlyextended the clinical phase and incubation periods ofprions. Interestingly, peripheral administration ofNPs-derived supernatants ameliorated the clinicalsigns associated to prion disease; however, did nothave effect on the incubation periods. Our findingssuggest that intracerebral andperipheral administra-tion of iPS-derived NPs might be potentially used totreat prion diseases.

PRION 73

138. Human cerebral organoids propagatesporadic CJD prions

Bradley R. Grovemana, Simote Foliakia, Christina D.Orrua, Gianluigi Zanussob, James A. Carrolla, BrentRacea and Cathryn L. Haigha

aLaboratory of Persistent Viral Diseases, National Institute of Allergy andInfectious Diseases, Division of Intramural Research, Rocky MountainLaboratories, National Institutes of Health, 903 South 4th Street,Hamilton, MT 59,840; bDepartment of Neurosciences, Biomedicine andMovement Sciences, University of Verona, Verona, Italy

CONTACT Cathryn L. Haigh [email protected]

ABSTRACT

Introduction: Human prion disease has traditionallybeen difficult to model in cell culture. Very fewhuman cell lines show sustained prion propagationover time and none represent three-dimensional (3D)brain tissue. Recently it was found that astrocyte cul-tures differentiated from human induced pluripotentstem cells (Hu-iPSCs) take up and propagate sporadicCJD (sCJD) prions with the resulting infection influ-enced by the sCJD subtype [1]. Hu-iPSCs have alsobeen utilized to differentiate self-patterning, structuredhuman brain tissues called cerebral organoids [2,3]. Wehypothesized that these cerebral organoids could beused to model human prion propagation in vitro.Materials and Methods: Cerebral organoid cultures weregenerated from hu-iPSCs using the protocol developed byLancaster and Knoblich [2]. Cerebral organoids wereinfected with one of two sCJD inocula and monitored forhealth, morphology, PrP seeding activity, PrP depositionand proteinase-k resistant PrP as compared with organoidsthat received a normal brain homogenate inoculum.Results: Cerebral organoids showed uptake and propa-gation of prions, detected by RT-QuIC analysis, follow-ing exposure to either sCJD inocula. Inoculum specificdifferences were observed. One inoculum showedrobust proteinase-K resistant PrP in all tested organoidsand coarse, granular PrP staining in the organoid inter-iors. The other inoculum induced a more toxic pheno-type that affected organoid health but displayed a lowlevel of prion seeding activity with no detectible pro-tease-resistant PrP.Conclusions: Cerebral organoid cultures can be used tomodel human prion disease in vitro. These structured 3Dmini-tissues offer a capacity to investigate aspects of priondiseases never previously available in a human neuronalcell model. Potential human disease-specific applicationsinclude delineating sCJD subtype pathogenesis and test-ing putative therapeutics.

KEYWORDS: Cerebral organoid; sporadic CJD;induced pluripotent stem cell; RT-QuIC

References

[1] Krejciova Z, Alibhai J, Zhao C, et al. Human stem cell-derived astrocytes replicate human prions in a PRNPgenotype-dependent manner. J Exp Med. 2017;214(12):3481–3495.

[2] Lancaster MA and Knoblich JA. Generation of cerebralorganoids from human pluripotent stem cells. NatProtoc. 2014;9(10):2329–2340.

[3] Renner M, Lancaster MA, Bian S, et al. Self-organizeddevelopmental patterning and differentiation in cere-bral organoids. Embo J. 2017;36(10):1316–1329

139. Cryo-electron microscopy of chronic wastingdisease prions

Sara Amidiana,b, Razieh Kamali-Jamila,b, Ester VázquezFernándeza,b, Maria Carmen Garzaa,b,$, Camilo DuqueVelásqueza,c, Xiongyao Wanga,b,#, Brian Tancownya,b,Chiye Kima, Judd Aikena,d, Debbie McKenziea,c,Howard Youngb and Holger Willea,b

aCentre for Prions and Protein Folding Diseases, University of Alberta,Edmonton, Canada; bDepartment of Biochemistry, University ofAlberta, Edmonton, Canada; cDepartment of Biological Sciences,University of Alberta, Edmonton, Canada; dDepartment of AnimalHealth and Biomedical Sciences, University of Alberta, Edmonton,Canada

CONTACT Sara Amidian [email protected]

Present address: $University of Zaragoza, Zaragoza, Aragón, Spain

Present address: #School of Materials Science and Engineering,Harbin Institute of Technology, Weihai, Shandong, China.

ABSTRACTIntroduction: Chronic wasting disease (CWD) is a fatalneurodegenerative disease in cervids that belongs to agroup of diseases known as prion diseases. CWD hasbeen detected in Canada, United States, South Korea,Norway, and most recently in Finland. CWD is the mostcontagious prion disease and poses a risk for transmissionto other species including humans.Aim: PrPSc is the misfolded form of the cellular prionprotein (PrPC) and the infectious agent that causes priondiseases. Themolecular mechanism of PrPC to PrPSc trans-formation is unknown. To understand this structural con-version, it is essential to understand the structure of PrPSc.Thus, the goal of this project is to investigate the structureof PrPSc forms that are causing CWD, using negative stainand cryo electron microscopy (EM).Method: PrPSc amyloid fibrilswere purified from the brainsof CWD-infected Tg33 mice, which express deer prion

74 ABSTRACT

protein. To purify PrPSc amyloid fibrils, phosphotungstateanions and sarkosyl were used. In addition, enzymes suchas proteinase K (PK) or pronase E (PE) were added toremove PrPC. To assess the quality and quantity of thePrPSc fibrils, the purified amyloid fibrils were negativelystained and visualized using EM. Cryo-EM and 3D fibrilreconstruction are then performed on fibril preparations.Results and Conclusion: PrPSc amyloid fibrils weresuccessfully purified and visualized using negativestain EM. Both, PK- and PE-purified samples displayedcomplex yet similar morphologies. However, a domi-nant and recognizable type seen in both preparationsconsisted of filaments with a clear twisted ribbon-likemorphology. 2D class averages and 3D reconstructionsfrom the negatively stained micrographs could help tobetter classify these fibrils. Additionally, cryo-EM ana-lyses could resolve the structural differences betweenconformers of distinct CWD prion strains and theirfibrillization properties. PE-purified fibrils, on theother hand, exhibited striations that run perpendicularto the fibril axis. Interestingly, the distance betweenthese striations is ~40 nm, which corresponds to themolecular height of two PrPSc molecules in a four-rungbeta solenoid architecture that may adopt a head-to-head arrangement [1]. This observation could beexplained by the presence of sarkosyl molecules asso-ciating with the hydrophobic GPI-anchor of the stackedPrPSc molecules. Further image processing analyses willprovide a more complete understanding of the orienta-tion of the GPI-anchor in the CWD fibrils.

References

[1] Wille H, Requena J. The Structure of PrPSc Prions.Pathogens. 2018;7:20. DOI:10.3390/pathogens7010020.

140. Molecular dynamics simulations of cervidprion protein variants to assess protein stabilityand susceptibility towards chronic wasting disease

Sara Amidiana,b, Lyudmyla Doroshc,d, Camilo DuqueVelásqueza,e, Maria Stepanovac,d, Judd Aikena,f,Debbie McKenziea,e and Holger Willea,b

aCentre for Prions and Protein Folding Diseases, University of Alberta,Edmonton, Canada; bDepartment of Biochemistry, University ofAlberta, Edmonton, Canada; cNational Institute for Nanotechnology,University of Alberta, Edmonton, Canada; dDepartment of Electricaland Computer Engineering, University of Alberta, Edmonton, Canada;eDepartment of Biological Sciences, University of Alberta, Edmonton,Canada; fDepartment of Animal Health and Biomedical Sciences,University of Alberta, Edmonton, Canada.

CONTACT Sara Amidian [email protected]

ABSTRACT

Introduction: Chronic wasting disease (CWD) is aprion disease that affects cervids. The central event inprion diseases is the structural conversion of thehealthy, cellular prion protein (PrPC) to the disease-causing isoform, PrPSc. It is technically difficult toexperimentally study the initial steps of the PrPC toPrPSc conversion; however, molecular dynamics (MD)simulation plays an important role in probing the mis-folding events. One approach is to assess the effect ofsingle-residue substitution on the structural stability ofPrPC. Polymorphisms in the prion protein gene caninfluence prion disease susceptibility, disease progres-sion, clinical presentation, and the propagation of dis-tinct CWD strains associated with different PrPScconformations. In this project, we use moleculardynamics simulation as a tool to characterize the effectof white-tailed deer (WTD) prion protein polymorph-isms Q95H and G96S on the experimentally deter-mined structure of the natively folded prion protein.Methods: The initial model for the WTD prion protein(residues 93–233) was generated using the crystal struc-ture of recombinant deer prion protein (PDB: 4YXH)as the template. The wild type (WT), 95H, and 96Smodels were subjected to minimizations, equilibrations,and production MD simulations using the Gromacspackage. In total, three independent 50ns simulationswere performed for each system.Results: The 96S polymorphism displayed a higher rootmean square deviation (RMSD) and root mean squarefluctuation (RMSF) values, which are indicators oflower structural stability compared to WT and 95Hforms. Also, 96S had a larger radius of gyration (Rg)values, indicating that the structure of 96S was lesscompactly folded than WT and 95H. Calculating thesolvent accessible surface area (SASA) of 96S revealedthat the structure of 96S was more exposed to thesolvent. On the other hand, structural dynamics of the95H polymorphism were closer to WT. RMSF and Rgvalues for 95H indicated higher stability and compact-ness of its structure. Additionally, the SASA values of95H were very similar to WT.Conclusions: Experimentally, the 96S allele is asso-ciated with slower disease progression and animalscarrying this polymorphism have longer incubationtime when inoculated with CWD prions. According toour MD results, it is apparent that the structure ofWTD prion protein carrying the 96S polymorphism isless stable than the WT protein. The clear difference inthe structural stability and local dynamics of 96S

PRION 75

compared to WT, may be relevant to its partial resis-tance to prion disease, however, further analyses arerequired.

141. Propagation of human sCJD prions inorganotypic slice culture

Grant Norman, Hailey Pineau and Valerie L. Sim

Department of Medicine, Division of Neurology, Faculty of Medicine& Dentistry, University of Alberta; Centre for Prions and ProteinFolding Diseases, University of Alberta

CONTACT Valerie L. Sim [email protected]

ABSTRACT

Prion diseases, or transmissible spongiform encephalo-pathies (TSEs), are neurodegenerative diseases that areinvariably fatal. These diseases result from the conver-sion of the normal prion protein (PrPC) to a misfoldedform (PrPSc) that can template itself and spreadthrough the brain, triggering neurodegeneration. TSEscan affect many mammals, including cervids (ChronicWasting Disease; CWD), sheep (scrapie), cattle (BovineSpongiform Encephalopathy; BSE), and humans(Creutzfeldt-Jakob Disease; CJD). There are no treat-ments for any of the prion diseases, despite decades ofresearch. One challenge to finding successful therapiesis the fact that PrPSc can exist in a number of differentconformations, each of which may have its own bio-physical properties and cause distinct neuropathologies,giving rise to unique prion strain phenotypes.Treatments that target only one specific conformationalchange in may only work for one strain. Ideally, ther-apy studies for humans should be done with humanprion strains. Here we demonstrate the propagation ofhuman prions in prion organotypic slice culture withevidence of associated neuronal loss. Traditionally,prion strains are generated in animals infected withprion disease; however, these are lengthy experiments,often taking several months. By contrast, the prionorganotypic slice culture assay (POSCA) can faithfullyrecapitulate aspects of prion pathology on a shortenedtimescale of 40–50 days [1,2]. This makes POSCA anideal method for propagating prion strains. Originally,POSCA was developed to test mouse-adapted scrapiestrains in cerebellar slices [2,3]. We have adaptedPOSCA to sporadic CJD strains from human samplesand samples passaged through mice expressing humanPrP. We have also adapted the system to whole braincoronal slices. The ability to propagate human prionstrains ex vivo will allow investigation of human strain-specific properties during pathogenesis as well as devel-opment of strain-specific therapeutic interventions.

Because POSCA is an open system, it also allows fortesting treatments at discrete disease time points with-out the confounder of blood-brain barrier.

References

[1] Falsig J, et al. Nat Protoc. 2008;3(4):555–62.[2] Falsig J, et al. PLoS Pathog. 2012;8(11):e1002985.[3] Campeau J, et al. PLoS One. 2013;8(12):e81776.

142. Effects of Elk-PrPC expression levels on CWDstrain properties

Camilo Duque Velásqueza, Alicia Oteroa, Chiye Kima,Elizabeth Triscotta, Jeffrey Narayana, Jacques Van derMerweb, Judd Aikenc and Debbie McKenziea

aDepartment of Biological Sciences, Centre for Prions and ProteinFolding Diseases, University of Alberta, Edmonton, Canada;bDepartment of Medicine, Centre for Prions and Protein FoldingDiseases, University of Alberta, Edmonton, AB, Canada;cDepartment of Agricultural, Food and Nutritional Sciences,Centre for Prions and Protein Folding Diseases, University ofAlberta, Edmonton, AB, Canada

CONTACT Alicia Otero [email protected]

ABSTRACT

Chronic Wasting Disease (CWD) is a contagious priondisease affecting various species of free-ranging and/orcaptive cervids on three continents. Species-specific prionprotein (PrPC) polymorphisms influence prion conver-sion into PrPCWD. PrPC amino acid variation can alsoregulate disease susceptibility to particular prion strainsand has been implicated in the diversification of prionstrain conformers [1, 2, 3]. Elk and deer PrPC differ atresidue E226Q and this amino acid difference has beenimplicated in the selection of CWD1 and CWD2 prionstrains [4]. As PrPC expression has been suggested toaffect prion strain evolution [5], we hypothesized thatelk PrPC levels affect CWD strain generation. To testthis hypothesis, transgenic (tg) FVB mice over-expressingelk PrPC [6] were crossed with prnp knock-out FVB miceto generate tg-elk with different PrPC expression levels.Both tg-elk+/+ and tg-elk± were exposed to white-taileddeer CWD strains (Wisc-1 and H95+) [2, 3]. The H95+

strain was a mixture generated on passage of Wisc-1 indeer heterozygous for H95G96 and Q95S96 [2]. Tg-elk+/+

mice succumbed to Wisc-1 with a mean incubation per-iod of 116 ± 7 days post infection (dpi) compared to 164 ±11 dpi for the H95+ strain mixture. Consistent with thereduced PrPC expression, the same deer prion strainsresulted in longer incubation periods (157 ± 21 dpi and>180 dpi, respectively) when passaged in tg-elk± mice.After first passage, transmission of Wisc-1 and H95+ in

76 ABSTRACT

tg-elk+/+ mice resulted in a single neuropathological pro-file that differed from the profile produced by passage ofelk prions (described as the CWD2 strain [1]). Our resultsshow that, upon first passage, the E226Q polymorphismdid not affect the strain properties of deer prions andindicates a single strain (Wisc-1) was selected by the tg-elk+/+ mice. The comparative analysis of neuropathologi-cal profiles between high and low expression tg-elk onfirst and second passage will be presented.

References

[1] Johnson CJ, Herbst A, Duque-Velasquez C, et al. Prionprotein polymorphisms affect chronic wasting diseaseprogression. PLoS ONE. 2011;6(3):e17450.

[2] Duque Velasquez C, Kim C, Herbst A, et al. Deer prionproteins modulate the emergence and adaptation ofchronic wasting disease strains. J Virol. 2015;89(24):12,362–12,373

[3] Herbst A, et al. Chronic wasting disease prion strainemergence and host range expansion. Emerg InfectDis. 2017;23(9):1598–1600.

[4] Angers R, et al. Prion strain mutation determined byprion protein conformational compatibility and pri-mary structure. Science. 2010;328, 5982, 1154–1158.

[5] Le Dur, et al. Divergent prion strain evolution drivenby PrPC expression level in transgenic mice. NatCommun. 2017;8:14,170.

[6] La Fauci et al. Passage of chronic wasting disease prioninto transgenic mice expressing Rocky Mountain elk(Cervus elaphus nelsoni) PrPC. J Gen Virol. 2006;87:3773–3780.

143. A role for molecular chaperones in the rate ofAlzheimer’s disease

Sarah C. Halesa, Hailey Pineaua,b, Grant Normana,b

and Valerie L. Sima,b

aCentre for Prions and Protein Folding Diseases, University ofAlberta; bDepartment of Medicine, Division of Neurology, Facultyof Medicine & Dentistry, University of Alberta

CONTACT Sarah C. Hales [email protected]

ABSTRACT

Despite similar pathological findings upon post-mor-tem examination of Alzheimer Disease (AD) brains(i.e. β-amyloid plaques and neurofibrillary tangles),the rate at which patients progress through this dis-ease is highly variable, ranging from less than 2 yearsin very rare cases, to an average of 10 years.Previously, studies seeking to understand heterogene-ity in disease progression have primarily looked fordifferences in the hallmark proteins associated with

AD: amyloid-β (Aβ) and tau. In terms of Aβ, studieshave found differences in Aβ plaque composition1and peptide structure2 in more rapidly progressingforms of the disease. Furthermore, the presence ofdifferent tau ‘strains’ has been suggested to influenceits ability to aggregate and spread.3 However, thestructural changes in Aβ and tau have not shedlight on why disease duration varies so dramaticallybetween patients. Our research into fast and slowcases of AD has revealed an increase in Aβ42 in themore slowly progressing cohort of patients, a findingthat was unexpected as Aβ42 is often thought to bethe more toxic species of Aβ. This may indicate thatother factors present in the cellular environment maydrive the rate of disease over and above any differ-ences in Aβ and tau. Given the established role formolecular chaperones in the regulation of proteosta-sis and mediation of protein aggregation, they areideal candidates to investigate with regards to rateof AD progression.

To test the role of chaperones in rate of disease,we are measuring levels of DNAJA2, HSP70, andHSC70 in human brains from patients with fastversus slow AD. We are also manipulating the levelsof chaperones to determine their effect on Aβ andtau aggregation in cell culture model systems (N2Acells containing the Swedish amyloid precursor pro-tein mutation, HEK293 cells containing the tauP301L mutation, and HEK293 cells containing therepeat domain of tau). A summary of these interac-tions and correlations will be presented.

References

[1] Drummond E, et al. Acta Neuropathol 2017; 133:933–954.

[2] Cohen ML et al. Brain 2015; 138:1009–1022.[3] Kaufman SK, et al. Neuron 2016; 92:796–812.

144. Deer prion protein polymorphisms influencein vitro aggregation kinetics

Helen Y. Yaoa, Leonardo M. Corteza,b, José Miguel Flores-Fernándeza,c, Holger Willea,c, Debbie McKenziea,d andValerie L. Sima,b

aCentre for Prions and Protein Folding Diseases, University of Alberta;bDepartment of Medicine, Division of Neurology, Faculty of Medicine &Dentistry, University of Alberta; cDepartment of Biochemistry, Faculty ofMedicine & Dentistry, University of Alberta; dDepartment of BiologicalSciences, Faculty of Science, University of Alberta

CONTACT Helen Y. Yao [email protected]

PRION 77

ABSTRACT

Introduction: Chronic wasting disease (CWD) is atransmissible neurodegenerative disease of cervids.It is caused by the misfolding and self-templatedaggregation of the prion protein (PrP). Differentpolymorphisms of PrP are associated with differentin vivo disease susceptibility, as shown in mice, elkand white-tailed deer [1]. Polymorphisms such as95H and 96S in deer Prnp are associated withaltered transmission properties, but the mechanismby which this happens is not clear. Deer heterozy-gous for the with G96S and/or Q95H polymorph-isms have longer incubation periods than deerexpressing wt Prnp [1]. Mice expressing 96S deerPrP are not susceptible to Wisc-1 strain CWD.Interestingly, PrP aggregates containing 95H areable to infect mice expressing 96S [2]. If thesetransmission differences are caused by differentaggregation and seeding efficiencies of the threepolymorphic PrP types, we may find similar correla-tions with in vitro aggregation studies: PrP contain-ing 96S or 95H should be less effective as a substratefor conversion, unless the seed contains 95H-PrP.Similarly, we expect any aggregate with 95H to bemore efficient than 96S as a seed.Methods: Wild type, 96S and 95H deer PrP wereexpressed in E. coli and purified from inclusion bodiesby affinity chromatography followed by HPLC.Monomeric PrP was aggregated using real-time quak-ing induced conversion (RT-QuIC) to study thekinetics of spontaneous, homologously seeded and het-erologously seeded aggregation. Lag phases and finalThT fluorescence values were compared.Results: As predicted, 96S was the most inefficient sub-strate for conversion, even when homologously seeded.Spontaneously formed aggregates of 96S were also theleast efficient seeds. 95H, on the other hand, producedaggregates that were very efficient at seeding, even morethan wildtype aggregates. 95H was also an excellent sub-strate for conversion, being more efficient than wildtype.Conclusions: The inefficient aggregation and seedingability of 96S PrP correlates with its long incubationperiod in vivo, suggesting this amino acid change couldexplain the in vivo findings. Similarly, the high seedingability of 95H PrP aggregates correlates with the findingthat 95H strains can infect otherwise resistance mice(96S mice). However, 95H was a very efficient substratein vitro which is not what has been observed in deerheterozygous for 95H; whether deer homozygous for95H would have faster incubation periods remains tobe seen.

References

[1] Johnson C, et al. J Gen Virol 2006;87:2109–114.[2] Duque Velasquez C, et al. J Virol 2015;89:12,362–73.

145. Deciphering ovine scrapie complexity inEurope by using bioassay in two rodent models

Alba Marín-Morenoa, Patricia Aguilar-Calvoa, José LuisPitarcha, Juan Carlos Espinosaa, Olivier Andreolettib,Romolo Nonnoc, Jan Langeveldd and Juan MaríaTorresa

aCentro de Investigación en Sanidad Animal (CISA-INIA), Madrid,Spain; bÉcole Nationale Vétérinaire de Toulouse, Toulouse, France;cIstituto Superiore di Sanitá, Rome, Italy; dWageningenBioVeterinary Research, Lelystad, the Netherlands.

CONTACT Alba Marín-Moreno [email protected]

ABSTRACT

Within the prion field, scrapie tends to be named as auniform disease caused by a single prion strain. However,as the opposite that applies for Bovine SpongiformEncephalopathy, several scrapie strains producing differentprion diseases phenotypes have been reported in the litera-ture. Previous work using European goat prion isolatesalready pointed to this situation (Nonno et al.). Goat andsheep scrapie isolates proceeding from several Europeancountries were intracranially inoculated into transgenicmice overexpressing ovine and bovine PrP proteins (Ov-Tg501 and Bo-Tg110 respectively). Two iterative passageswere performed. The combination of the two differentrodent models allows the classification of the isolates infour groups that may correspond to several scrapie strains.Abrogation of the transmission barrier by performing thesecond passage is necessary to correctly classify the isolates.In addition, Bo-Tg110 provides the most important infor-mation to allow discrimination between groups.

KEYWORDS: Scrapie; prion strain; transmission bar-rier; Europe; mouse bioassay.

Funding

Spanish Ministerio de Ciencia, Innovación yUniversidades (AGL2016-78054R) and EuropeanUnion projects CT-2006–36,353 and 219,235 ERA-NET FP7 EMIDA.

146. Conversion kinetics of sporadic CJD prionsusing multiple substrates and RT-QuIC-baseddetection and discrimination

Stephanie Bootha, Sarah Medinab, Lise Lamoureuxb

and Debra Sorensenc

78 ABSTRACT

aPHAC; bPublic Health Agency of Canada; cPublic Health Agency ofCanada-NML

ABSTRACT

Prions often maintain their conformational characteristicsupon natural transmission to a new species and that this isdependent on the structural characteristics of both the‘infecting’ PrP and the new host PrP. Based on the hypoth-esis that variations in prion conformation are intrinsic tostrain characteristics and ultimately disease phenotypes, itseems logical to conclude that the seed – substrate pairinghas a direct relationship to the conversion rate in assayssuch as real-time quaking-induced conversion (RT-QuIC).We have used RT-QuIC to assess the relative rates ofconversion of CJD prions from over 40 patients forwhom CJD was confirmed as the cause of death by theCanadian CJD Surveillance System. These patientsincluded those diagnosed with five subtypes of sporadicCJD, sCJDMM1/sCJDMV1, sCJDVV2, sCJDMV2,SCJDMM2 and sCJDVV1. A number of different recom-binant prion proteins including hamster, elk and humanwere used as substrate. Serial dilution of brain tissue fromthese individuals were used as seeds. We hypothesized thatthe degree of complementarity during seed-substrate pair-ing would provide a measure of strain conformation basedon the relative efficiency of the initial amyloid conversionof a particular substrate by a seed. We measured the con-version rates by comparing the time at which fluorescencein each reaction exceeded a pre-defined threshold, i.e. thelag phase. Lag phases were extremely consistent for eachCJD type in the different substrates so that we couldstatistically compare cases and assign to different types.Of interest, we found a small number of atypical caseswhose conversion kinetics differed significantly fromothers within their subtype perhaps indicating differentstrain types. We also performed seeding studies on sCJDprions isolated from specific brain regions of interest pro-viding information on the heterogeneity of prions relatedto the brain region in which they are propagated.

147. Novel, pan-PrPSc-specific antibodies reveal ashared β-solenoid structure for all PrPSc strainstested

Holger Willea,b, Andrew Fanga,b, Xinli Tanga,b, MadeleineFleminga,b, Tolu Abodunwaa,b, Leonardo M. Corteza,c,Claudia Y. Acevedo-Morantesa,b, Camilo DuqueVelásqueza,d, Brian Tancownya,b, Xiongyao Wanga,b,$,Judd Aikena,e, Debbie McKenziea,d and Valerie L. Sima,c

aCentre for Prions and Protein Folding Diseases, University ofAlberta; bDepartment of Biochemistry; cDepartment of Medicine;dDepartment of Biological Sciences; eDepartment of Agricultural,Food, and Nutritional Science, Edmonton, Alberta, Canada.

CONTACT Holger Wille [email protected]

$current address: School of Materials Science andEngineering, Harbin Institute of Technology, Weihai,Shandong, China.

ABSTRACT

Background: The high-resolution structure of PrPSc

has eluded experimental determination due to its inso-lubility and its general propensity to aggregate.Nevertheless, the repeating nature of PrPSc amyloidfibrils has allowed the collection of lower-resolutionstructural data via X-ray fibre diffraction, cryo-electronmicroscopy, and other techniques. All of theseapproaches indicated that the structure of PrPSc isbased on a four-rung, parallel β-solenoid architecture.Materials and Methods: The aforementioned β-solenoidarchitecture allowed us to predict which amino acid sidechains may be oriented towards the surface of PrPSc

versus those that form the interior of the β-solenoidfold. Based on these predictions, we created a structuraland immunological mimic for PrPSc by inserting out-wards facing residues into an identical position in aninnocuous protein scaffold based on the fungal prionHET-s. This structural and immunological mimic (=vac-cine candidate) was then injected into wild-type mice andtheir immune response analysed. The resultant spleno-cytes were used to generate hybridoma cells for the pro-duction of monoclonal antibodies. These monoclonalantibodies were characterized for their ability to recognizenative PrPSc. Lastly, a systematic series of revertantmutants of the PrPSc-based vaccine candidate towardsthe unmodifiedHET-s scaffold were used to further refinethe epitopes that these antibodies recognize.Results: Immunization with the structure-based vaccinecandidate resulted in a PrPSc-specific immune response in8 out of 8 mice. The post-immune antiserum recognizedonly native PrPSc, and did not bind to PrPC, recombinantPrP, denatured PrP, or any linear PrP peptides. Themonoclonal antibodies that were derived from the immu-nized mice (IgGs and IgMs) recognized all natural PrPSc

strains/isolates, including BSE, CWD, TME, scrapie, andhuman prions (familial: fCJD, FFI, GSS; sporadic: sCJD;infectious: vCJD). Moreover, the monoclonal antibodiesdid not recognize in vitro generated recPrP amyloid,which presumably adopted a parallel in-register intermo-lecular β-sheet (PIRIBS) architecture. Lastly, the HET-srevertant mutants helped to define the PrPSc epitopeswithin the framework of the HET-s β-solenoid structure.Conclusions: Our novel, pan-PrPSc-specific antibodiesconfirm the hypothesis that the structure of PrPSc con-tains a parallel β-solenoid fold. In particular, theseantibodies recognize structural epitopes that are

PRION 79

specific for the parallel β-solenoid fold of PrPSc, but donot recognize alternative conformers of PrP includingβ-sheet rich PIRIBS structures. These antibodies andthe original antigen are currently being studied for theirpotential use as passive or active immunotherapyagents and for use in diagnostic applications.

148. Characterization of infectious human prions

Claudia Y. Acevedo-Morantesa,b, Xiongyao Wanga,b,*,Brian Tancownya,b, Susana Teijeirac,d, Beatriz SanMillánc,d and Holger Willea,b

aCentre for Prions and Protein Folding Diseases; bDepartment ofBiochemistry, University of Alberta, Edmonton, AB, Canada; cGaliciaSur Health Research Institute, IIS Galicia Sur, SERGAS-UVIGO, Vigo,Spain; dPathology Department, Complexo Hospitalario Universitariode Vigo (CHUVI), IIS Galicia Sur, SERGAS, Vigo, Spain.

CONTACT Claudia Y. Acevedo-Morantes [email protected]

*Current address: School of Materials Science andEngineering, Harbin Institute of Technology, Weihai,Shandong, China.

ABSTRACT

Human prion diseases are a family of rare and progressiveneurodegenerative disorders. They present as sporadic,familial, infectious, or iatrogenic forms, and includeCreutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome (GSS), and Fatal Familial Insomnia(FFI). The causes for the wide range of phenotypic varia-tion in the human prion diseases are unknown, but thedisease is caused by aberrantly folded versions of the prionprotein, termed PrPSc. Biochemical properties such aspartial resistance to digestion by proteinase K, insolubilityin non-ionic detergents, and sedimentation properties haveprovided evidence that distinct forms of PrPSc aggregateswith heterogeneous morphologies regarding fibril length,width, shape, longitudinal twist, and the number of proto-filaments that may characterize these disease entities. Inthis study, we are using several biochemical and biophysicalapproaches to analyse and compare PrPSc-fractionsderived from the brains of patients affected by sporadicCJD (sCJD), familial CJD (fCJD), variant CJD (vCJD), GSS,and FFI. It is hypothesized that the structure of PrPSc is thekey factor that determines its infectious nature, transmis-sion route, and pathogenicity. However, the insolubility ofPrPSc and its overall propensity to aggregate make it inac-cessible to high-resolution structure analyses. Low-resolu-tion approaches have provided information on thearchitecture of PrPSc to devise molecular models. Usingtilted-beam transmission electron microscopy (TB-TEM),we are measuring the mass-per-length (MPL) value ofindividual PrPSc fibrils derived from the brains of patients

affected by the aforementioned prion diseases. Theseresults will provide a platform to evaluate the proposedmodels for the structure of PrPSc fibrils. Tobacco MosaicVirus (TMV), with its well-determined structure and anMPL value of 131 kDa/nm, serves as an internal calibrationstandard for the MPL measurements. Preliminary resultsobtained with purified PrPSc fibrils from sCJD samplesindicated a population of fibrils with a MPL value around60 kDa/nm, whichwould support the four-rung β-solenoidmodel based on two protofilaments per fibril. AdditionalMPL measurements on GSS patient-derived samples areunderway. Recombinant PrP amyloid fibrils as well as invitro converted recombinant PrPSc will be used to roundout the MPL comparison of prion fibril structures. MPLdata impose a stringent constraint on viable fibril modelswhen the subunit mass is known. By calibrating the imageintensity of PrPSc to the internal standard, we will be ableto limit the number of possible indexing schemes signifi-cantly, and provide independent experimental validation tothe current proposed models.

149. Rationally designed, structure-based vaccinescandidates for aβ, tau and α-synuclein

José M Flores-Fernández, Enrique Chimal-Juárez,Xiongyao Wang*, Brian Tancowny, Xinli Tang,Andrew Fang and Holger WilleCentre for Prions and Protein Folding Diseases & Department ofBiochemistry, University of Alberta, Edmonton, Alberta, Canada.

CONTACT José M Flores-Fernández [email protected]

*current address: School of Materials Science andEngineering, Harbin Institute of Technology, Weihai,Shandong, China.

ABSTRACT

Background: Alzheimer’s and Parkinson’s diseases aremainly caused by the misfolding of Aβ, tau, and α-synu-clein proteins, respectively. Each of these proteins adopts abeta-sheet rich conformation and aggregates into amyloid.Different attempts to develop vaccines for these diseaseshave used the respective proteins and peptides in theirlinear form and without control of their tertiary and qua-ternary structure, which caused a lack of specificity andsubsequently a failure to achieve protection. In this study,we present a new approach to design vaccine candidatesbased on structurally defined proteins that result in disease-specific antigenicity.Materials and Methods: The fungal prion Het-s wasused as an innocuous scaffold protein because itnatively adopts a conformation rich in beta-sheet.This scaffold protein was engineered based on the sur-face exposed amino acids of Aβ, tau, and α-synuclein

80 ABSTRACT

aggregates to express antigenic determinants in a struc-turally controlled manner. The resulting vaccine candi-dates were produced in Escherichia coli, purified,refolded, and then analysed by EM for the properfolding of the proteins. Only those constructs thatformed amyloid fibrils with the typical structure ofHet-s amyloid were injected into mice. The resultingpolyclonal antibodies were tested against unmodifiedHet-s, the engineered Het-s antigen, and disease-speci-fic antigens in order to determine their specificity.Results: After the purification and refolding of theengineered proteins, several vaccine candidates thattargeted Aβ, tau, and α-synuclein showed the expectedself-assembly into amyloid fibrils by negative stain elec-tron microscopy. The self-assembled constructs wereinjected in mice and the collected antisera were testedfor their specificity by ELISAs using Het-s and itsengineered variants as antigens. The vaccine candidatesAβ4 and Tau4 elicited immune responses with higherreactivity for the engineered antigens than for unmodi-fied Het-s.Conclusions: Rationally designed, structure-based vac-cine candidates targeting the misfolded proteinsresponsible for Alzheimer’s and Parkinson’s diseaseare feasible with this approach.

150. Characterization of the glycosylation profileof water-soluble prion protein from brain andblood of hamster using lectins

Hanin Abdel-Haq

Istituto Superiore di Sanità, Rome, Italy

CONTACT Hanin Abdel-Haq [email protected]

ABSTRACT

Background: The disease-associated water-soluble formof hamster prion protein (ws-PrPSc) has recently beenfound to be less stable than classical PrPSc. Since thestability of PrP to enzymatic degradation correlates withits glycosylation level, the aim of this study was to inves-tigate whether there are differences between the glycosy-lation of ws-PrPSc and classical PrPSc of hamster.Materials and methods: To achieve this aim, ws-PrP andclassical PrP were captured from noninfected or scrapie-infected hamster brain homogenate [high-speed superna-tant (SHS) and high-speed pellet (PHS)] and blood plasmaby anti-PrP antibodies (3F4 and 6H4) and subjected toscreening for glycans by seven biotinylated lectins underdenaturing or nondenaturing conditions in a sandwichlectin enzyme-linked immunosorbent assay (lectin-ELISA).

Results: Glycans have been found in minor quantitiesand differently exposed on ws-PrPSc from SHS andplasma compared with classical PrPSc from PHS.These differences have been shown to be potentiallyresponsible for the instability of ws-PrPSc. Treatmentof infected blood with guanidine hydrochloride(GdnHCl) significantly (P <0.01) increased the detec-tion of ws-PrPSc in ELISA, reflecting an increase inits stability, and showed efficacy in removing high-abundance proteins in silver-stained gels. Thisincrease in ws-PrPSc stability is due to an interactionof GdnHCl not only with high-abundance proteinsbut also with the ws-PrPSc glycosylation with parti-cular regard to the mannose sugar. Analysis of lectinsimmunoreactivity towards total proteins from plasmacollected before and at different time points afterinfection, revealed that mannose might exert a stabi-lizing effect towards all of hamster blood glycopro-teins, regardless of scrapie infection. Since low levelsof ws-PrPSc/soluble-infectivity have been estimatedboth in blood and brain of hamster, this glycosyla-tion-related instability may have negatively influ-enced the propensity of ws-PrPC to convert to ws-PrPSc both in blood and the brain.Conclusions: Glycosylation characteristics of cellularprion protein (PrPC) may provide a tool for the determi-nation risk of prion transmissibility among different spe-cies and tissues. These results could be representative forother misfolded proteins that are characterized by havingdifferent glycoforms, including the soluble form, such asamyloid beta in Alzheimer’s disease.

KEYWORDS: Glycosylation; Prion protein;Guanidine-induced protein stabilization; Lectins;Blood; Misfolded proteins.

151. Early detection of prion protein aggregationwith pFTAA using spectral confocal microscopy

Waqas Tahira, Anastasiia Stepanchukb, Hermann MSchatzla and Peter K Stysb

aDepartment of Comparative Biology and Experimental Medicine,Faculty of Veterinary Medicine, University of Calgary, Calgary,Alberta, Canada; bDepartment of Clinical Neuroscience, HotchkissBrain Institute, University of Calgary, Calgary, Alberta, Canada

CONTACT Waqas Tahir [email protected]

ABSTRACT

Background: Prion diseases are fatal neurodegen-erative diseases characterized by the formation ofamyloidogenic prion aggregates in the CNS. Prionaggregates are formed by conformational alterationof host encoded cellular prion protein into the

PRION 81

scrapie form. Quick progression in disease pathol-ogy after the onset of symptoms makes such diseasesdeadlier. Early detection of prion aggregates canhelp to better understand the pathophysiology andspread of prion infection in the brain. In thisregard, amyloid sensitive fluorescent probes andadvanced analytical tools to quantify emission spec-tra of those fluorescent probes can help to detectthese pathological events at earlier stages of diseaseprogression.Objective: Early detection of protein aggregates inprion infected mouse brains by examining changes inemission spectrum of brain sections stained with theamyloid-sensitive fluorescent probe pFTAA by usingspectral confocal microscopy.Materials and Methods: FVB mice were intra-cerebrallyinoculated with 20 μL of brain homogenates (mock or22 L infected) from terminal mice. Prion infected andage matched mock inoculated mice were euthanized forpre-symptomatic stages (at 75 and 100 dpi), sympto-matic stage (125 dpi) and terminal stage. Brain tissueswere extracted and fixed with formalin. Brain sections(5 μm) were hydrated followed by incubation withpFTAA (3 µM) for 45 min. Stained sections were imagedwith confocal microscope equipped with a 32-channelspectral detector. Image analysis was done in custom-written software ImageTrak using a non-linear spectralunmixing algorithm.Results: Fluorescence spectral analysis of prion-infected brain sections from pre-symptomatic stages(75 and 100 dpi) showed increased fluorescenceintensity as well as a characteristic red shift in theemission spectrum as compared to age matchedmock brain sections. Differences in emission spec-trum between prion infected and mock inoculatedbrain sections increased at later stages of the dis-ease. Spectral differences at pre-symptomatic stageswere mainly seen in somatosensory cortex, hippo-campus and thalamus. Pathological alterationsrepresented by changes in emission spectrum duringsymptomatic and terminal stages spread to furtherbrain regions including olfactory bulb, hypothala-mus, cerebellum, and brainstem.Conclusions: Advanced spectral imaging analysis ofpFTAA stained brain sections infected with 22 Lprions showed robust differences in emission spec-trum during prion infection. This shows the potentialof such methods for early detection of prion aggre-gates, possibly also in the tissues other than onlybrain, and also can offer better understanding aboutmechanism of prion spread in protein misfoldingdisorders with higher sensitivity.

152. Tissue tropism of prions is dictated by thebalance between PrPSc formation and degradation

Ronald A. Shikiyaa, Katie A. Langenfeldb, Thomas E.Ecklandc, Jonathan Trinha, Sara A. M. Holeca, Candace K.Mathiasond,e, Anthony E. Kincaida,e and Jason C. Bartza

aDepartment of Medical Microbiology and Immunology, CreightonUniversity, Omaha, NE, USA; bBiology Department, University ofNebraska at Omaha, Omaha, NE, USA; cDepartment of Neurology,University of Texas Health Science Center, Houston, TX, USA;dDepartment of Microbiology, Immunology and Pathology,Colorado State University, Fort Collins, CO, USA; eDepartment ofPharmacy Science, Creighton University, Omaha, NE, USA

CONTACT Ronald A. Shikiya [email protected]

ABSTRACT

Prion strains are characterized by strain-specific differ-ences in neuropathology, incubation period, clinical dis-ease, host-range, and tissue tropism. In hamsters, thehyper (HY) and drowsy (DY) prion strains differ intissue tropism and susceptibility to disease when extra-neural routes of infection are used. Notably, hamstersusceptibility to the DY strain is restricted when inocu-lation occurs via extraneural routes. In contrast to theHY strain, DY is not detected in secondary lymphoreti-cular system (LRS) tissues of infected hosts regardless ofthe route of infection. Our experiments show that, simi-lar to the lymphotropic strain HY, DY crosses the muco-sal epithelia, enters the draining lymphatic vessels inunderlying laminae propriae, and is transported to LRStissues. Since DY is able to cause disease once it entersthe peripheral nervous system, the restriction in DYpathogenesis is not due to a failure of transport butmay be due to its inability to stablish infection in LRStissues. To determine if DY can propagate in LRS tissues,we performed protein misfolding cyclic amplificationusing DY PrPSc as the seed and spleen homogenate asthe source of PrPC. Our results show that the spleenenvironment can support DY PrPSc formation, albeit atlower levels when compared to other lymphotropicprion strains, suggesting that the restriction in DYpathogenesis is not due to the absence of a strain-specificco-factor in spleen. In addition, DY PrPSc is more sus-ceptible to degradation compared to PrPSc from otherlymphotropic strains. Based on these results, we suggestthat the equilibrium between PrPSc formation and clear-ance plays a role in prion tissue tropism.

153. Molecular mechanisms of Aβ fibril elongation,secondary nucleation, and fibril dissociationstudied in-silico

Lyudmyla Dorosha,b and Maria Stepanovab,c

82 ABSTRACT

aDepartment of Electrical and Computer Engineering, University ofAlberta, Edmonton, Canada; bNational Institute for NanotechnologyNRC; Edmonton, Canada; cDepartment of Electrical and ComputerEngineering, University of Alberta, Edmonton, Canada

CONTACT Lyudmyla Dorosh [email protected]; MariaStepanova [email protected]

ABSTRACT

Prevalence in ageing population, difficulties in early diag-nosis, and lack of efficient therapies make Alzheimer’sdisease (AD) one of the most devastating neurodegenera-tive disorders of our time. AD is associated withmisfoldingand formation of toxic aggregates of amyloid β (Aβ) pep-tide, subsequently producing β-rich amyloid deposits [1].Evidence has emerged that amyloid fibrils may catalyse theprocess of corruptive misfolding through a positive-feed-back reaction loop [2]. However, detailed molecularmechanisms of this process remain elusive. In an effort tobetter understandmolecular mechanisms of Aβmisfoldingand fibrillization, we performed all-atom moleculardynamics (MD) simulations of interaction of Aβ1-42 chainswith fragments of β-rich amyloid fibrils in water. As thefibril models, we employed the solution NMR model2NAO [3] and the cryo-EM model 5OQV [4]. Four dis-ordered Aβ1-42 chains were placed in random positions at adistance from the fibrils, and evolution of the entire systemswas investigated. Multiple independent simulations weredone for each system. For each fibril model, we observedattachment of Aβ1-42 chains to the fibril. Importantly, boththreading of Aβ1-42 chains around the edge of the fibril(precursor of fibril elongation) and attachment of thechains to fibril’s side surface (precursor of secondarynucleation) were detected with each of two fibril models.For the 5OQV model, threading around both the ‘ridge’and ‘groove’ ends of the fibril were observed. Surprisingly,attachment of Aβ1-42 chains to side surface of 5OQV fibrilsoccurred with the Aβ chains’ preferential orientation alongthe fibril axis, rather than along cross-β strands. In one ofsimulations with the 2NAO fibril model, we observeddetachment of an Aβ chain caused by its interaction witha different Aβ chain in solution (precursor of fibril dissocia-tion). In contrast, the 5OQV fibrils have exhibited aremarkable structural stability in our simulations. In con-clusion, the MD simulations have shed light into earlystages of two major molecular mechanisms of misfoldingand aggregation of Aβ1-42, fibril elongation and secondarymisfolding, as well as captured a fibril dissociation event,revealing important molecular-level details behind theseprocesses.

References

[1] Eisenberg & Jucker, Cell 2012; 6:1188–1203.

[2] Cohen et al., PNAS 2013; 110:9758–9763.[3] Walti et al., PNAS 2016; 113:E4976-E4984.[4] Gremer et al., Science 2017; 358:116–119.

154. TSEs in European goats discriminated byWestern blotting into five types based on fiverobust molecular parameters

L Pirisinua, O Andreolettib, I Lantierc, PL Acutisd, C Acine,W Goldmannf, T Sklaviadisg, L Ekateriniadouh, C Fasti,P Papasavva – Stylianouj, S Simonk, J Spiropou-losl, U Agrimia, JG Jacobsm, A Bossersm, LJM vanKeulenm, M Mazzad, R Nonnoa and JPM Langeveldm

aIstituto Superiore di Sanità, Department of Food Safety, Nutrition andVeterinary Public Health, Rome, Italy; bUMR INRA ENVT 1225- IHAP, ÉcoleNationale Vétérinaire de Toulouse, Toulouse, France; cINRA-Centre Valde Loire, Infectiologie et Santé Publique, Nouzilly, France; dIstitutoZooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta,Torino, Italy; eCentro de Encefalopatías y Enfermedades TransmisiblesEmergentes, Facultad de Veterinaria, Universidad de Zaragoza,Zaragoza, Spain; fThe Roslin Institute and Royal (Dick) School ofVeterinary Studies, University of Edinburgh, Easter Bush, UnitedKingdom; gLaboratory of Pharmacology, School of Health Sciences,Department of Pharmacy, Aristotle University of Thessaloniki,Thessaloniki, Greece; hNational Agricultural Research Foundation,Veterinary Research Institute, Thessaloniki, Greece; iInstitute of Noveland Emerging Infectious Diseases, Friedrich-Loeffler-Institute,Greifswald-Isle of Riems, Germany; jVeterinary Services, Nicosia, Cyprus;kService de Pharmacologie et Immunoanalyse (SPI), Laboratoired’Etudes et de Recherches en Immunoanalyse, CEA, INRA, UniversitéParis-Saclay, Gif-sur-Yvette, France; lAnimal and Plant Health Agency,New Haw, Addlestone, Surrey, United Kingdom; mWageningenBioVeterinary Research, Lelystad, the Netherlands

CONTACT Laura Pirisinu [email protected]

ABSTRACT

Contrasting the knowledge about prion diseases orTSEs in sheep, only a very limited number of straintyping studies are available in goats. Two cases deriv-ing from the zoonotic bovine BSE epidemic werehowever detected in goats. During 2004–2012, over70 TSE goat brain samples were collected from sevenEuropean countries and evaluated for TSE type/strainvariation. A selection of these materials was chosenfor in-depth analysis based on various criteria: tissuequality, genotype, broad geographical distribution,potential type variation. Of these, 37 cases were bio-chemically analysed (present study) and a subset ofthese subjected to bioassays in seven rodent models(see parallel study of Nonno et al.). Analyses of theseisolates showed that in goats different PrPSc typesexist, comparable to those found in sheep such asclassical scrapie, CH1641-like scrapie and Nor98/aty-pical scrapie. However, classical scrapie isolates couldbe further differentiated in two molecular subtypesbased on the PK susceptibility of the N-terminus and

PRION 83

on the molecular weight of PrPres. Importantly, thetwo subtypes of classical scrapie showed differentgeographical distribution, as one subtype was onlydetected in goats deriving from Italy and France.These analyses also show, that none of the fieldsamples exhibited BSE-like features and offer a com-prehensive set of robust molecular PrPres propertiesto exclude suspicion for the presence of BSE. Theseare: molecular mass of the triplet bands, presence ofN-terminus epitope, glycoprofile markers, a dualpopulation marker and the structural stability ofPrP-core.

This work was made possible by national support andby the following European grants: Neuroprion (FP6-FOOD-506,579), GoatBSE (FOOD-CT-2006–36,353) andGOAT-TSE-FREE (EMIDA-ERA NET). We want tomemorize Jorg G Jacobs, who died in 2017. Jorg developedand performed the Triplex-WB system as well as charac-terized antibodies like 94B4, 12B2, 9A2, 6C2, SAF84 andL42. He also was crucial in the distinction of C-, L- and H-type BSE and in 2011 he showed elegantly the allotypecomposition of prion material in heterozygous sheep bythe use of Endo-LysC.

155. Expert opinions on the potential role ofIndigenous peoples in wildlife management inAlberta

Arlana Bennett

University of Alberta

CONTACT Arlana Bennett [email protected]

ABSTRACT

Background: Natural resource management in manyparts of the world are increasingly based around therecognition of Indigenous knowledge and the rights ofresource users [1]. In Alberta, there has been a histor-ical lack of consideration of the knowledge of FirstNations and Métis peoples and decisions about thedevelopment, use, management and monitoring ofnatural resources [2–5]. As a result of these histories,Indigenous peoples and their knowledge have littleinfluence within provincial wildlife managementregimes in contemporary Alberta [6]. Wildlife diseasessuch as Chronic Wasting Disease (CWD) have addedfurther challenges for wildlife managers in Alberta.CWD is a fatal form of transmissible spongiformencephalopathy (TSE) primarily found in cervids (e.g.deer, moose, elk, and caribou). CWD in Alberta as ofthe 2017/2018 hunting season, has continued tospread westward along the Red Deer/South

Saskatchewan/Bow watershed, the Battle watershed,and north along the Alberta/Saskatchewan border.Incidence of deer that have tested positive for CWDhas increased markedly since the 2016/2017 huntingseasons, and several new Wildlife Management Units(WMU) will be added to the mandatory surveillancelist for the 2018/19 hunting season [7]. First Nationsand Métis communities in the Treaty 6 area – includ-ing Saddle Lake First Nation, Frog Lake First Nation,Kehewin First Nation, Fishing Lake Métis settlement,and the Elizabeth Métis settlement – are within themandatory surveillance zone yet it remains unclearwhether these communities are being actively engagedin the process of management.Methods: This research sought to explore diverse expertperspectives on the role of Indigenous Knowledge in wild-life monitoring and management in relation to the issue ofCWD; and better understand the key challenges andopportunities regarding wildlife management in Alberta.The methods used in this thesis include a modified quali-tative expert elicitation, probabilistic sampling, and the-matic analysis.Results: The major thematic results experts discussedinclude: the lack of Indigenous compliance in cervidmonitoring with varying reasons provided; the neces-sity of both scientists and Indigenous communities toengage in intercultural and technical capacity devel-opment; and the need for both scientists andIndigenous communities to form a functional andmutually beneficial working relationship. Thisresearch is a preliminary investigation into the social,cultural, and economic aspects of CWD manage-ment, and is intended to provide further insightstowards this end with a focus on future areas ofresearch.

References

[1] Berkes F. Sacred Ecology. 2018.[2] Sandlos J. Hunters at the margin. 2007.[3] Loo T. States of nature. 2006.[4] Fumoleau R. As long as this land shall last. 2004.[5] Calliou BL. Losing the game. 2000.[6] Natcher, et al. Applied Anthropology. 2009;68:245–

257.[7] AEP (Alberta Environment and Parks). CWD Updates.

2018.

156. Screening and characterization of unusualsCJD cases in a CWD endemic state in the USA

Yihui Liua, Manuel Camachoa, Wenquan Zoua,b,c,Qingzhong Konga,b,c

84 ABSTRACT

aDepartment of Pathology, Case Western Reserve University (CWRU),Cleveland, USA; bDepartment of Neurology, CWRU, Cleveland, OH, USA;cNational Center for Regenerative Medicine, CWRU, Cleveland, USA

CONTACT Qingzhong Kong [email protected]

ABSTRACT

Background: Chronic wasting disease (CWD) hasspread to 26 states in the USA and three provinces inCanada, and it has been detected recently in Norwayand Finland. Potential CWD zoonosis is a serious pub-lic health concern. It is unclear whether CWD trans-mission to humans has already occurred. We aim tostart to address this question by examining all availablesCJD cases from a CWD endemic state in the USA.Methods: Frozen brain tissues from all available sCJDcases archived in the National Prion Disease PathologySurveillance Center from a US state that has beensignificantly impacted by CWD were sampled at fivebrain regions. These brain samples were subjected todetailed biochemical analysis to look for unusual pat-terns, characteristics, and/or distribution of PrPSc incomparison with sCJD samples from states that havenot detected CWD. Unusual cases are further scruti-nized for their clinical presentations, histopathologicalfeatures, and history of cervid hunting and venisonconsumption.Results and Conclusions: We have found some unu-sual sCJD cases in this CWD endemic state. We willreport our preliminary findings on their features.Currently there is no convincing evidence to supporta direct link to CWD for any of these unusual sCJDcases.Disclaimer: The findings and conclusions in this reportare those of the authors and do not necessarily repre-sent the position of the National Prion DiseasePathology Surveillance Center.

157. Utility of RT-QuIC in predicting case status ofpotential Creutzfeldt-Jakob Disease (CJD) cases

Natalie S. Marzeca, Marie-Ange Smithb, Jennifer A.Housea

aColorado Department of Public Health and Environment, Denver, CO,USA; bColorado School of Public Health, Aurora, CO, USA

CONTACT Natalie S. Marzec [email protected]

ABSTRACT

Background: Ante-mortem diagnosis of human priondisease is limited by the high false positivity rate of thetesting methods available for cerebrospinal fluid (CSF).This has resulted in a lack of efficiency when

investigating potential prion disease cases for surveil-lance purposes. In 2018, the real time quake-inducingconversion (RT-QuIC) test was added to the CDC diag-nostic criteria for Creutzfeldt-Jakob Disease (CJD), themost common prion disease reported in humans. RT-QuIC appears to be a better predictor of Definite orProbable CJD case status than the older Tau and 14-3-3 protein tests. The purpose of this project was to eval-uate the utility of RT-QuIC to predict case status ofpotential CJD cases.Methods: Potential cases of CJD reported in Coloradobetween 1 January 2015, and 31 December 2017 (priorto the change in the CDC diagnostic criteria), werereviewed for final case status, testing methods, andante-mortem test results. RT-QuIC, Tau, and 14-3-3test results were compared to final case status to deter-mine the positive predictive value (PPV), negative pre-dictive value (NPV), sensitivity, and specificity of eachtest. Cases that met either Definite or Probable casedefinitions were considered to be true cases.Results: Sixty-six potential cases of CJD reported in thespecified time period were investigated and assigned a casestatus of Definite, Probable, Possible, or did not meet casedefinition. Of these cases, 27 (41%) were assigned aDefinite or Probable case status. RT-QuIC was found tohave a PPV of 92.8%, a NPV of 100%, a sensitivity of 100%and a specificity of 95.8%. Tau protein had a PPV of 48.6%,a NPV of 93.8%, a sensitivity of 94.7%, and a specificity of55.9%. 14-3-3 protein had a PPV of 32.4%, aNPVof 83.3%,a sensitivity of 92.3%, and a specificity of 16.7%.Conclusions: RT-QuIC testing is a more useful ante-mortem predictor of Definite or Probable CJD case statusthan either Tau or 14-3-3 protein testing. Public healthinvestigators can place more weight on RT-QuIC testingthan other ante-mortem CJD disease testing when tria-ging cases for investigation. To our knowledge, this is thefirst time RT-QuIC testing has been studied in relation topredicting public health surveillance case status.

KEYWORDS: Creutzfeldt-Jakob disease; RT-QuIC;public health; surveillan

158. Familial Creutzfeldt-Jakob disease withD178N and Met129Val

Nurit Omera, Esther Kahanab, Sharon Simhonic, AnatBar-Shirac, Tova Naimanc, Avi Orr-Urtregerc, NirGiladid and Noa Bregmana

aCJD clinic, Neurological institute, Tel Aviv Sourasky Medical Center,Tel Aviv, Israel; bDepartment of Neurology, Barzilai Medical Center,Ashkelon, Israel; cThe Genetic Institute, Tel-Aviv Sourasky MedicalCenter, Tel-Aviv, Israel; dNeurological institute, Tel Aviv SouraskyMedical Center, Tel Aviv, Israel

PRION 85

CONTACT Nurit Omer [email protected]

ABSTRACT

Fatal familial insomnia (FFI) and Creutzfeldt-Jakob disease(CJD) are two phenotypes that share a common pointmutation at codon 178 of the prion protein gene (PRNP),with the substitution of aspartic acid to asparagine(D178N). The phenotype depends on the polymorphismat codon 129, when methionine (129M) is associated withFFI, and Valine (129V) with CJD. We here report on aJewish family from Iraqi origin with D178N-129V geno-type presenting with overt insomnia, cerebellar ataxia, andan atypical duration of disease. Patient 1, a 56-year-oldwoman without family history of neurological illness, pre-sented in 1996 with rapidly progressive dementia and gaitataxia. MRI showed frontal cortical but not basal ganglia orthalamic involvement. CSF was positive for protein 14-3-3,and genetic testing for E200Kmutation was negative. Brainbiopsy showed typical spongiform changes, and she wasdiagnosed with sporadic CJD. She passed away 7 years aftersymptoms onset. Her son (patient 2) presented on 2012with rapidly progressive dementia at the age of 41 and wasdiagnosed with CJD. He died a fewmonths later. Patient 3,her daughter, presented on 2018 at the age of 50, withcerebellar ataxia and mild cognitive impairment. Shedescribed severe insomnia for more than a year beforearrival. Brain MRI showed overt restriction in diffusionacross the frontal, parietal and temporal cortices, withmarked involvement of basal ganglia and thalami. A geneticanalysis of the PRNP gene revealed a heterozygous D178Nmutation (sequence variant: c.532G>A) in combinationwith the polymorphism Met129Val (sequence variant:c.385A>G) on both alleles (homozygous). Patient 4, herthird daughter, 45 years old, is currently (2019) undergoingmedical evaluation due to reported progressive cognitivedecline for 6 months. The phenotype of patient 1 wasclearly different from that of FFI and more suggestive ofCJD, although the long duration of the disease is highlyunusual for CJD. Severe insomnia was reported in the‘prodromal’ phase in patient 3, with marked thalami invol-vement in MRI, but with a genetic signature that is con-sistent with CJD. Our findings support the notion thatthere is a phenotypic variability in the D178N genotypewithin the same family, and emphasize the heterogeneity ofinherited prion disease and the possibility that genetic andother modifiers exist.

159. Therapeutic and clinical strategy to preventor delay onset of genetic prion disease using prionprotein-lowering antisense oligonucleotides

Eric Vallabh Minikela,b,c, Hien Tran Zhaod, Deb Cabine,Stuart L. Schreibera,f, Jeffrey B. Carrolle, Holly

Kordasiewiczd,g, Steven E. Arnoldb,h, Byron Caugheyi

and Sonia M. Vallabha,b,c

aBroad Institute of MIT and Harvard, Cambridge, MA, USA; bHarvardMedical School, Boston, MA, USA; cPrion Alliance, Cambridge, MA, USA;dIonis Pharmaceuticals Inc, Carlsbad, CA, USA; eMcLaughlin ResearchInstitute, Great Falls, MT, USA; fHarvard University, Department ofChemistry and Chemical Biology, Cambridge, MA, USA; gWesternWashington University, Bellingham, WA, USA; hMassachusetts GeneralHospital, Department of Neurology, Boston, MA, USA; iLaboratory ofPersistent Viral Diseases, Rocky Mountain Laboratories, NationalInstitute of Allergy and Infectious Diseases, National Institutes ofHealth, Hamilton, MT, USA

CONTACT Sonia M. Vallabh [email protected]

ABSTRACT

Background: Genetic proofs of concept suggest thatreduction of prion protein (PrP) in the brain should bean effective and well tolerated therapeutic strategy forprion disease. We are therefore investigating PrP-lower-ing antisense oligonucleotides (ASOs) as a potential priondisease therapeutic. Based on initial observations of ASOefficacy in prion-infected mice (see complementaryabstract by Dr Byron Caughey) here we assess parametersof ASO efficacy including dose responsiveness, strainspecificity, and impact of treatment timepoint. As earlytreatment conveys greater benefit, we also describe aframework for testing PrP-lowering therapeutics in pre-symptomatic prion disease mutation carriers, includingbiomarker development, establishment of a carrier cohortand regulatory engagement.Materials and methods: All mice received bolus dosesof active ASOs or saline by stereotactic intracerebro-ventricular injection, then were monitored for symp-toms with a primary outcome of terminal prion disease.ASO doses, prion strains and treatment initiation time-point varied across experiments. To evaluate CSF PrPas a biomarker for PrP-lowering therapeutics, N = 217human CSF samples spanning a range of diagnoses andprion disease subtypes were subjected to PrP quantifi-cation by ELISA.Results: ASO efficacy is dose responsive, universal acrossall prion strains tested, and more efficacious if adminis-tered early. In an ongoing experiment, PrP-lowering ASOswere administered every 90 days beginning at −14 dpi; allsaline-treated animals have reached prion endpoint (mean143 dpi) while all but one ASO-treated animal is living at316 dpi, corresponding to a ≥ 2.2x extension of survival.CSF PrP is detectable in all human CSF samples tested,reliably quantifiable by ELISA subject to proper handling,appears CNS-derived, and exhibits excellent short-termwithin-subject test-retest reliability (mean CV = 8.5%) ingenetic prion disease mutation carriers, suggesting thatASO-mediated PrP lowering should be readily detectablein the context of a trial. In a recent Critical Path Innovation

86 ABSTRACT

Meeting, we discussed with FDA scientists a path to pre-symptomatic trials in genetic prion disease mutation car-riers based on CSF PrP lowering as surrogate endpoint forprovisional approval of an ASO.Conclusions: These data support advancement of PrP-lowering ASOs to the clinic, and support the feasibilityof primary prevention trials in healthy genetic priondisease mutation carriers. We are working closely withour pharmaceutical partner, Ionis Pharmaceuticals, aswell as with FDA to advance ASOs to the clinic withinthe next few years. Genetic prion disease is poised tobecome a model for primary prevention inneurodegeneration.

160. Rapid bioassay of mammalian prions inDrosophila

Alana M. Thackray and Raymond Bujdoso

Department of Veterinary Medicine, Cambridge University,Cambridge, UK

CONTACT Raymond Bujdoso [email protected]

ABSTRACT

Mammalian prions cause fatal neurodegenerative diseasessuch as Creutzfeldt-Jakob disease (CJD) in humans, BovineSpongiform Encephalopathy (BSE) in cattle, chronic wast-ing disease (CWD) in cervids and scrapie disease of sheep.These conditions are transmissible between individuals ofthe same or different species and as a consequence, animalprion diseases pose a realistic threat to human healththrough their zoonotic potential. Prions lack a conventionalnucleic acid-based genome. The ‘protein only’ conceptpredicts that infectious prion particles consist of PrPSc inthe form of aggregates of misfolded conformers of thenormal host protein PrPC, although the exact molecularnature of this transmissible entity is undefined. As a con-sequence, the only reliable method to detect mammalianprion infectivity is by bioassay in an appropriate experi-mental host. It is important to be able to detect prioninfectivity in tissues and fluids from individuals affectedby prion disease in order to protect human and animalhealth. In addition, it is important to address the zoonoticpotential of animal prion infectivity in order to ensure thesafety of the human food chain. The detection of prioninfectivity by bioassay in mammalian species, most com-monly mice, is time consuming and expensive, and conse-quently, lownumbers of samples are routinely assessed.Wehave previously demonstrated Drosophila can bioassaymammalian prions (Thackray, et al. Brain 2018;141:2700–2710). Our studies have shown that ovine PrPtransgenic Drosophila develop a transmissible neurotoxicphenotype, associated with Proteinase-K resistant PrPSc,

after exposure to exogenous ovine prions.We have recentlygenerated Drosophila transgenic for human, bovine or cer-vid PrP and have demonstrated that these flies can be usedsuccessfully to bioassay variant CJD, BSE or CWD samples,respectively. In addition, we have shown that human PrPtransgenic Drosophila can be used to model the zoonoticpotential of animal prions. Collectively, our novel datashow that Drosophila can detect human and animal prioninfectivity in a rapid and efficient manner, and can con-tribute to an understanding of the zoonotic potential ofanimal prion disease.

161. Rodent models allow BSE discrimination ofgoat prions and reveal geographical differences inthe biological properties of scrapie

R. Nonnoa, A. Marin-Morenob, J.C. Espinosab, C. Fastc, L.Van Keulend, J. Spiropoulose, I. Lantierf, O. And-reolettig, L. Pirisinua, M.A. Di Baria, P. Aguilar-Calvob,T. Sklaviadish, P. Papasavva-Stylianoui, P.L. Acutisj, C.Acink, A. Bossersd, J.G. Jacobsd, G. Vaccaria, C. D’Ag-ostinoa, B. Chiappinia, F. Lantierf, M. Groschupc, U.Agrimia, J.M. Torresb and J.P.M. Langeveldd

aIstituto Superiore di Sanità, Department of Food Safety,Nutrition and Veterinary Public Health, Rome, Italy;bCentro de Investigación en Sanidad Animal, CISA-INIA,Madrid, Spain; cInstitute of Novel and Emerging InfectiousDiseases, Friedrich-Loeffler-Institute, Greifswald-Isle ofRiems, Germany; dWageningen BioVeterinary Research,Lelystad, the Netherlands; eAnimal and Plant HealthAgency, New Haw, Addlestone, Surrey, United Kingdom;fINRA-Centre Val de Loire, Infectiologie et Santé Publique,Nouzilly, France; gUMR INRA ENVT 1225- IHAP, ÉcoleNationale Vétérinaire de Toulouse, Toulouse, France;hLaboratory of Pharmacology, School of Health Sciences,Department of Pharmacy, Aristotle University ofThessaloniki, Thessaloniki, Greece; iVeterinary Services,Nicosia, Cyprus; jIstituto Zooprofilattico Sperimentale delPiemonte, Liguria e Valle d’Aosta, Torino, Italy; kCentrode Encefalopatías y Enfermedades TransmisiblesEmergentes, Facultad de Veterinaria, Universidad deZaragoza, Zaragoza, Spain

CONTACT Romolo Nonno [email protected]

ABSTRACT

Classical scrapie (CS) is a contagious TSE of smallruminants known to circulate in Europe for centuries.More than one TSE strain is responsible for CS, still ourability to identify scrapie strains remains limited, as ithas been difficult so far to reconcile data obtained indifferent rodent models. After the discovery of BSE intwo goats, a European-broad effort was undertaken in

PRION 87

order to understand the diversity of goat TSE strainsand to discriminate BSE from them. Here, we studiedgoat TSEs by their relative transmission efficiency indifferent animal models and by the PrPSc type(s) pro-pagated in rodents. Thus, the biological properties ofTSE isolates were inferred by their specific interactionwith different recipient PrP species rather than by serialpassage in a single model. Goat TSE isolates from sevencountries and experimental goat-BSE were inoculated inseven models: tg-mice overexpressing sheep/goat ARQ,sheep VRQ, cattle or mouse PrPs; RIII mice and bankvoles. The attack rate and the survival time were com-bined to obtain the ‘transmission efficiency’, a para-meter used to build the ‘transmission profiles’ ofindividual isolates across all models. Goat-BSE andNor98 showed distinctive transmission profiles, differ-ent among them and from all other isolates. Goat-BSEwas the only isolate inducing 19kDa PrPSc in all rodentmodels, while Nor98 was associated with 8kDa PrPSc.CS isolates showed a strong variability of transmissionprofiles, which was in part explained by their geogra-phical distribution, and could be grouped into fourdistinct categories. CS isolates in category 1 inducedthe propagation solely of 21kDa PrPSc, while isolatesfrom CS categories 2-to-4 also induced, to a differentextent depending on the model, a 19kDa PrPSc typedistinct from BSE. Further investigation of CNS andLRS tissues from a goat herd showed that this phenom-enon reflects the selective amplification in rodents of atleast two co-existing ‘sub-strain components’, 21kDaand 19kDa, whose relative amount varies with differentgoat tissues/isolates. Our findings show that BSE can bediscriminated from a wide range of biologically andgeographically diverse goat TSEs. The geographical var-iation observed in CS suggests the existence of distinctCS strains, as also supported by the parallel studies byPirisinu et al. and Marin-Moreno et al. Yet, CS isolatesin categories 2-to-4 were mixtures of discrete sub-strains, where it could be envisaged that competitionamong sub-strains play a role in defining the biologicalproperties of individual isolates and their evolution.

162. ‘Multi-species anti-prion oral vaccine forfamilial and transmissible spongiformencephalopaties’

Fernando Goñia, Analia Eliseib, Lucia Yimc, MitchellMarta-Arizaa, Jose A Chabalgoityc, Thomas Wisniewskia,d

aDepartment of Neurology New York University, NY, USA; bINTA,Buenos Aires Argentina; cBiotech Department of Universidad de laRepublica, Uruguay; dDepts of Pathology and PsychiatryNYUSoM, USA

CONTACT Fernando Goñi [email protected]

ABSTRACT

Background/Introduction: Transmissible prion diseaseshave become a worldwide problem of enormous propor-tions with no prevention or cure in sight. The NorthAmerican and northern Europe Chronic Wasting disease(CWD) epidemic in free range moose, elk and deer has notbeen contained. We were successful in developing an oralvaccination with PrP molecules delivered by an attenuatedSalmonella carrier to prevent transmission and infection ofsusceptible mice, and achieve, in white-tailed deer, partialmucosal and systemic antibody protection to an oral chal-lenge with an infective CWD-prion [1,2]. We have nowrefined the constructs to small peptideswith specificities formultiple mammalian PrP molecules for a more effectiveoral vaccine.Materials and Methods: We designed and constructed20–30 residues long multi-species PrP-like peptides withamino acids specific for at least three different species andencompassing the whole PrP molecule. Peptides wereexpressed in an attenuated Salmonella delivery system, orcontrolled glutaraldehyde polymerized with and withoutcholera toxin-B (CTB). Vaccine preparations were kept for7 days at 4°C and then for 36 h at RT. All preparations wereinoculated by gavage into mice transgenic for differentspecies PrP. After three weekly inoculations antibody pro-duction was measured in faeces and serum.Results: Viability of the vaccines after 10 days wasbetween 87% and 95%. Using a reduced number ofinoculations, all Tg CD-1 mice expressing human, elk,sheep or mouse PrP and PrP-KO produced a sustainedantibody response (both IgA and IgG) that cross-reacted with prion proteins from different species.Conclusions: Stability and ease of production of thesenew vaccine formulations would allow for up scaling anduse in larger animals of any species administered as foodsupplement in enclosed facilities or potentially as bait inthe wild. The cross-reactivity of the antibodies producedwould help neutralize any infective prion from oraltransmission and inhibit spread of the β-sheet structureresponsible for PrPC to PrPSc conversion [3].

KEYWORDS: CWD; pan-vaccine; multi-species; oralvaccine

References

[1] Goñi, et al. Neuroscience 2008; 153:679686.[2] Goñi, et al. Vaccine 2015; 33:726733.[3] Wisniewski and Goñi. Handbook of Clinical

Neurology 2018; 153:419–430.

88 ABSTRACT

163. Structural characterization of α-synucleinprions via cryo-electron microscopy

Gregory E. Merza, Eric Tsea, Victor Banerjeea, AmandaL. Woermana,b, William F. DeGradoa,d, Daniel R.Southwortha,c and Stanley B. Prusinera,b,c

aInstitute for Neurodegenerative Diseases, Weill Institute forNeurosciences, University of California, San Francisco, CA, USA;bDepartment of Neurology, University of California, San Francisco,CA, USA; cDepartment of Biochemistry and Biophysics, University ofCalifornia, San Francisco, CA, USA; dDepartment of PharmaceuticalChemistry, Cardiovascular Research Institute, University of California,San Francisco, CA, USA

CONTACT Gregory E. Merz [email protected]

ABSTRACT

Recent structural characterization via cryo-electronmicroscopy (cryo-EM) has confirmed at high-resolu-tion that different fibrillar structures of tau prions (ortau strains) exhibit clinically distinct tauopathy disor-ders. Such findings are consistent with the hypothesisthat the biological properties of prion strains are deter-mined by their misfolded conformations. Similarly,several groups have recently demonstrated that thesynucleinopathies of Parkinson’s disease (PD) andmultiple system atrophy (MSA) are caused by α-synu-clein aggregates with distinct biological activities.Given that the structure of prions determines theirbiological properties, we hypothesize that α-synucleinprions in PD and MSA have unique fibrillar conforma-tions. We are investigating this structural hypothesisby characterizing α-synuclein prions at high resolutionusing cryo-EM. Several strains of α-synuclein prionswill be purified from human brain as well as homo-genates passaged through Tg mice and cultured cells.We will perform cryo-EM on proteins purified frombrain homogenates to obtain the high-resolution infor-mation necessary to make structural comparisonsbetween strains at the molecular level. The overallfolds of prions, as well as inter- and intra-molecularinteractions, will be evaluated across prion strains.Mutation sites in α-synuclein causing familial synuclei-nopathies (e.g. A53T) are of particular interest, and theinteractions of these residues within the larger fibrillarfold will be closely studied. The unequivocal assign-ment of specific α-synuclein conformations with theresulting disease is likely to represent a major stepforward in our understanding of the pathogenesis ofsynucleinopathies.

164. Overview of scrapie cases in Iceland 2004–2018

Eva Hauksdottir and Stefania Thorgeirsdottir

Keldur, the Institute for Experimental Pathology, University ofIceland

CONTACT Eva Hauksdottir [email protected]

ABSTRACT

Introduction: Since 1978 Iceland has actively screenedhealthy slaughtered (HS) sheep for scrapie, however, in2004 the screening method changed from histopatho-logical staining to a rapid ELISA test. With this newmethod scrapie cases in HS were identified for the firsttime, as before index scrapie cases had only beendetected in sheep with clinical symptoms (CS). Use ofthis sensitive method also led to the detection of thefirst atypical scrapie case (Nor98) in Iceland in 2004.Materials and Methods: During 2004–2018 there were50,515 HS samples tested with ELISA, along with 558samples from fallen stock and clinical suspects. In thisoverview, the cases detected during this time, will beanalysed further, e.g. origin, age, PrP genotype andnumber of additional cases from each farm detectedafter culling of the scrapie flock.Results: In the years 2004–2018 there were 29 scrapiecases identified, where one case refers to a singleaffected farm. Two thirds of the classical scrapie casesoriginate from CS sheep, while Nor98 is generallyidentified in HS samples rather than CS. The genotypemost associated with susceptibility to classical scrapie isfrequent in the classical scrapie cases, however a neutralgenotype is found in many of those cases as well. Afterculling, where a scrapie flock (or a portion of it) wastested, additional positive samples of classical scrapiewere frequently detected, but only once has an addi-tional Nor98 case been found in a culled flock.Conclusion: The comparison of classical and atypicalscrapie in Iceland shows that they have many differentqualities in relation to origin, symptoms, genotype andflock infectivity.

165. Familial Parkinson’s point mutation abolishesmultiple system atrophy prion replication

Amanda L. Woermana,b, Sabeen Kazmia, Smita Patela,Abby Oehlera, Kartika Widjajaa, Daniel A. Mordesc,Steven H. Olsona,b and Stanley B. Prusinera,b,d

aInstitute for Neurodegenerative Diseases, Weill Institute forNeurosciences, University of California, San Francisco, CA, USA;bDepartment of Neurology, University of California, San Francisco, CA,USA; cC.S. Kubik Laboratory for Neuropathology, Department ofPathology, Massachusetts General Hospital, Boston, MA, USA;dDepartment of Biochemistry and Biophysics, University of California,San Francisco, CA, USA

CONTACT Amanda L. Woerman [email protected]

PRION 89

ABSTRACT

In the neurodegenerative disease multiple system atrophy(MSA), α-synuclein misfolds into a self-templating con-formation to become a prion. To compare the biologicalactivity of α-synuclein prions in MSA and Parkinson’sdisease (PD), we developed nine α-synuclein-YFP celllines expressing point mutations responsible for inheritedPD. MSA prions robustly infected wild-type, A30P, andA53T α-synuclein-YFP cells, but they were unable toreplicate in cells expressing the E46K mutation.Coexpression of the A53T and E46K mutations wasunable to rescue MSA prion infection in vitro, establish-ing that MSA a-synuclein prions are conformationallydistinct from the misfolded α-synuclein in PD patients.This observation may have profound implications fordeveloping treatments for neurodegenerative diseases.

166. α-cleavage of PrPC by Aspirin suppressesprion infection

Huyen Trang Trinha, Taeyeon Kimb, A-Ran Kima,Junwoo Shina, Hakmin Leea, Jieun Kima, SungeunLeea, Chongsuk Ryoua

aDepartment of Pharmacy, College of Pharmacy and Institute ofPharmaceutical Science and Technology, Hanyang University, Ansan,Gyeonggi-do, Republic of Korea; bDivision of Developmental Biologyand Physiology, School of Biosciences and Chemistry, Institute for BasicScience, Sungshin University, Seoul, Republic of Korea

CONTACT Huyen Trang Trinh [email protected];Chongsuk Ryou [email protected]

ABSTRACT

Background: The conversion of the cellular prion protein,PrPC, into its pathogenic form, PrPSc, is the hallmark ofprion diseases. In physiological conditions, PrPC can becleaved at α-site, which generates the C1 andN1 fragments,and disrupts a region critical for conversion into PrPSc.Plasmin is known as an enzyme that cleaves PrPC at α-site;however, the function of plasmin in prion propagationremains elusive. In the current study, the role of plasminand Aspirin, known to stimulate plasmin activity, in α-cleavage of PrPC and prion suppression was investigated.Material and Methods: The effect of plasmin andAspirin was evaluated using an in vitro assay, whichmimics the aggregation process of prion protein intoamyloids. In the cell-based model, ScN2a cells wereincubated with Aspirin and endogenous plasmin activ-ity was measured. The generation of C1 and N1 frag-ments were also detected by immunoblotting. Theelimination of PrPSc by Aspirin was investigated inScN2a. Finally, the ability of Aspirin to interfere with

the susceptibility of N2a to prions was determinedusing standard scrapie cell assay.

Results: Aspirin promoted plasmin activity by indu-cing α-cleavage of PrP, resulting in the inhibition ofPrP aggregation in vitro. Moreover, the endogenousplasmin activity in the ScN2a was significantlyenhanced by the action of Aspirin. The generation ofC1 fragment was increased in the membrane fractionof the cells incubated with Aspirin, indicating thatAspirin activated α- cleavage of PrPC. In addition,Aspirin significantly reduced the level of PrPSc incells permanently infected with prions. Finally,Aspirin significantly decreased the number of N2acells infected with prions.Conclusions: Aspirin promoting plasmin-mediated α-cleavage of PrPC suppressed PrPSc propagation.

KEYWORDS: Aspirin; plasmin; α-cleavage; C1fragment

167. Two different conformers of type 1 prionprotein propagate as distinct strains in transgenicmice

Ignazio Calia,b,* Juan Carlos Espinosac,*, TetsuyukiKitamotod, Alba Marin-Morenoc, Brian S. Applebya,b,e,Juan Maria Torresc and Pierluigi Gambettia

aDepartment of Pathology and bNational Prion Disease PathologySurveillance Center (NPDPSC), Case Western Reserve University,School of Medicine, Cleveland, OH 44,106, USA; cCentro deInvestigación en Sanidad Animal, CISA-INIA, Madrid, Spain;dDepartment of Neurological Science, Tohoku University GraduateSchool of Medicine, Sendai, Japan; eDepartment of Neurology,University Hospitals Cleveland Medical Center, Beachwood, OH44,122, USA.

CONTACT Ignazio Cali [email protected]

*These authors contributed equally to this work

ABSTRACT

A modern classification recognizes five subtypes of spora-dic Creutzfeldt-Jakob disease (sCJD)-based on the pairingof methionine (M)/valine (V) polymorphic genotype atcodon 129 of the prion protein (PrP), which determinestheMM,MV and VV 129 genotypes, and the type 1 or 2 ofthe disease-associated PrP (PrPD)1. These two PrPD typesare distinguished on the basis of their different electro-phoretic mobility as the PK-resistant PrPD (resPrPD) type1 and type 2 migrate to ~21 and ~19 kDa, respectively. InsCJDVV1, the electrophoretic profile of resPrPD is distinctfrom that of resPrPD type 1 associated with the sCJDMM(MV)1 subtype. It features a heterogeneous component,which in its unglycosylated form appears in three versions:a single band of ~20 kDa (T120), of ~21 kDa (T121) or as a

90 ABSTRACT

doublet with both ~20 and ~21 kDa bands (T120−21).Although these bands are a minor component resPrPD,their consistent presence raises the issue as to whether theyare a feature that distinguish the PrPD strain associatedwith sCJDVV1 from that associated with sCJDMM(MV)1and other type 1 strains. We have carried out a retrospec-tive examination of brain homogenates from subjects withthe definitive diagnosis of sCJDVV1 that were collected atthe National Prion Disease Pathology Surveillance Center(NPDPSC). Western blot (WB) analysis confirmed thepresence of the three distinct electrophoretic profiles ofunglycosylated resPrPD. To determine whether these dis-tinct molecular signatures of resPrPD associated withsCJDVV1 could be propagated in vivo, brain homogenatesfrom sCJDVV1 cases harbouring each of the three profileswere inoculated to transgenic (Tg) mice expressing humanPrP-129V. The T120 and T121 WB profiles were faithfullyreplicated in the Tg mice. Although as in sCJDVV1 theywere associated with undistinguishable histopathologicalphenotypes the incubation periods following T120 andT121 inoculations differed. Moreover, similar inoculationsto Tgmice expressing PrP-129M, i.e. mismatched at codon129 with the PrP-codon 129 of the inoculum, hamperedtransmission of T121 and delayed that of T120. Combined,these data indicate that T121 and T120 represent two dis-tinct human prion substrains which however are not ableto determine distinct phenotypes.

Funding

Supported by National Institutes of Health Grants R01NS083687 and P01 AI106705, and The Charles S.Britton Fund (to P.G.). This study was also supportedby a grant from the Spanish Ministerio de Ciencia,Innovación y Universidades (AGL2016-78054-R –AEI/FEDER, UE).

References

[1] Parchi et al. Ann Neurol. 1999;46(2):224–233.[2] Parchi et al. Acta Neuropathol. 2011;121:91–112

168. Clinical prediction of type 1 and type 2 mixedhistopathology distribution in MM-type sporadicCreutzfeldt-Jakob disease

Toshimasa Ikedaa,b, Yasushi Iwasakia, Keita Sakuraic, AkioAkagia, Maya Mimuroa, Hiroaki Miyaharaa, TetsuyukiKitamotod, Noriyuki Matsukawab, Mari Yoshidaa

aDepartment of Neuropathology, Institute for Medical Science ofAging, Aichi Medical University, Nagakute, Japan; bDepartment ofNeurology and Neuroscience, Nagoya City University GraduateSchool of Medical Sciences, Nagoya, Japan; cDepartment ofRadiology, Teikyo University School of Medicine, Tokyo, Japan;

dDepartment of Neurological Science, Tohoku University GraduateSchool of Medicine, Sendai, Japan

CONTACT Toshimasa Ikeda [email protected]

ABSTRACT

Background/Introduction: Although the sporadicCreutzfeldt-Jakob disease (sCJD) subtypes, with their dis-tinctive clinicopathological features, have been identifiedbased on two types of the abnormal prion protein (PrPSc),type 1 and type 2, and polymorphic codon 129, there havebeen cases of both PrPSc types being present. The mixedPrPSc type features, especially inMM-type CJD, have beenbest identified with neuropathological examination; theMM1-type involves fine vacuole-type spongiform change(FV), whereas the MM2C-type involves large confluentvacuole-type spongiform change (LCV). Presently, it isimpossible to detect the concurrence of both PrPSc typesbefore death. The purpose of this study was to clinicallypredict the concurrence of MM-type sCJD with anotherPrPSc type in the same individual.Materials and Methods: We retrospectively identifiedseven MM-type sCJD cases with both FV and LCVamong 49 sCJD cases genetico-pathologically diagnosedin our institute between 2008 and 2016. We reviewedclinical features, pathological findings, and radiologicalabnormalities. We also conducted a regional systemicstudy to associate the spongiform change pattern withhyperintensity on magnetic resonance diffusion-weightedimaging (DWI) using the signal intensity index (SII).Results: The one patient with dominant LCV showedlonger disease duration, later onset of typical symptoms,no periodic sharp wave complexes in electroencephalo-graphy, and negative 14–3-3 protein findings comparedto the six FV-dominant patients. LCV-dominant lesionstended to show higher intensity on DWI than FV-domi-nant lesions in respective patients. In the regional sys-temic study, LCV-dominant regions showed significantlyhigher SII on DWI than did the FV-dominant regions.Conclusions: Mixed MM-type sCJD showed clinicalfeatures correlating with the dominant pathologicalburden. The SII may be clinically useful for investigat-ing the concurrence of PrPSc type 2 in cases with thetypical clinical course of MM1-type sCJD.

KEYWORDS: Creutzfeldt-Jakob disease; histopathol-ogy; diffusion-weighted imaging (DWI); magnetic reso-nance imaging (MRI); spongiform change

169. Stable propagation of hamster prions inCRISPR/Cas9-engineered CAD5 cells

Matthew Bourkasa,b, Hamza Arshada,b, GeroldSchmitt-Ulmsa,c, Jason Bartzd and Joel Wattsa,b

PRION 91

aTanz Centre for Research in Neurodegenerative Diseases, Toronto,Canada; bDepartment of Biochemistry, University of Toronto, Toronto,Canada; cDepartment of Laboratory Medicine and Pathobiology,University of Toronto, Toronto, Canada 4 Department of MedicalMicrobiology and Immunology, Creighton University, Omaha, USA

CONTACT Matthew Bourkas [email protected]

ABSTRACT

Background:Prion research has been hindered by a lackof cellular paradigms for studying the replication ofprions from different species. Although hamster prionshave been widely used to study prion replication inanimals and within in vitro amplification systems, theyhave proven challenging to propagate in cultured cells.Since the murine catecholaminergic cell line CAD5 isuniquely susceptible to a diverse range of mouse prionstrains, [1,2] we hypothesized that it might also be cap-able of propagating non-mouse prions.Materials and Methods: We generated CAD5 cellslacking endogenous PrPC (CAD5-PrP−/- cells) usingCRISPR/Cas9-mediated genome editing. CAD5-PrP−/-

cells were stably transfected with a plasmid encodinghamster PrP and then challenged with brain homoge-nates containing various strains of hamster prions.Results: When exposed to the 263K, HY, or 139H strains,hamster PrP-expressing CAD5-PrP−/- cells stably propa-gated high levels of protease-resistant PrP. Moreover,when these cells were challenged with 10-fold serial dilu-tions of 263K prions, PK-resistant PrP was obtained withdilutions up to and including 0.000002% brain homoge-nate. Cellular homogenates from 263K- infected cellsexhibited prion seeding activity in the RT-QuIC assayand were infectious to naïve cells expressing hamsterPrP. The presence of endogenous mouse PrP in hamsterPrP- expressing CAD5 cells completely blocked the repli-cation of hamster prions. Interestingly, murine N2a neu-roblastoma cells ablated for endogenous PrP expressionwere susceptible to mouse prions, but not hamster prionsupon expression of cognate PrP.Conclusions: Our results demonstrate that CAD5 cells,but not N2a cells, can propagate prion strains fromnon-mouse species following the ablation of endogen-ous mouse PrP. This suggests that CAD5 cells eitherpossess cellular factors that enhance or lack factors thatrestrict the diversity of strains that can be propagated.We conclude that transfected CAD5-PrP−/- cells may bea useful tool for assessing the biology of prion strainsand dissecting the mechanism of prion replication.

References

[1] Mahal et al., Proc Natl Acad Sci USA. 2007;104(52):20,908–13.

[2] Berry et al., Proc Natl Acad Sci USA. 2013;110(44):E4160–9.

170. Biochemical characterization of PrPSc speciesat pre-clinical stages of disease in a mouse modelof prion disease (RML)

Ghazaleh Eskandari-Sedighia,b, Leonardo Corteza,c,

Jing Yanga, Hristina Gapeshinaa, Nathalie Daudea,Valerie Sima,c and David Westawaya,b,d

aCentre for prions and protein folding diseases, University of Alberta,Edmonton, Alberta, Canada; bDepartment of Biochemistry, Faculty ofMedicine and Dentistry, University of Alberta, Edmonton, Alberta,Canada; cDepartment of Medicine, Division of Neurology, Faculty ofMedicine and Dentistry, University of Alberta, Edmonton, Alberta,Canada; dDepartment of Medicine, Faculty of Medicine and Dentistry,University of Alberta, Edmonton, Alberta, Canada

CONTACT Ghazaleh Eskandari-Sedighi [email protected]

ABSTRACT

Background: It is known that the amount of PrPSc andinfectious titre in brains of infected animals can plateauat sub-clinical stages, suggesting a division of prioninfectivity and toxicity phases [1]. Previous work inour lab has shown that a post-translational downregu-lation of PrPC could account the plateau effect [2,3].These data also suggested that diminishing levels ofPrPC (i.e. the substrate for PrPSc formation), as wellas the rise in accumulated PrPSc are important deter-minants in the transition from sub-clinical to clinicalstage. Although transcriptomic studies point at majorrole of inflammation in prion diseases [4], and a recentstudy on global metabolomics in mice points at changesin brain chemistry at a mid-stage in disease progression[5], there is still ambiguity about the causal relationshipbetween these changes, neuronal loss and clinical man-ifestation of the disease.Material and Methods: Using mice inoculated with theRML isolate of mouse-adapted scrape, we applied differenttechniques to track the aforementioned parameters and thebiochemical features of the PrPSc particles. The isotropicfractionator technique [6] was used to quantify the numberof neural and non-neural cells in the brain of infectedanimals. Asymmetric-flow field- flow fractionation (AF4),a one-phase chromatography technique that separates par-ticles based on their size, was used to isolate the PrPScparticles from brains; size of the isolated particles wasdetermined by dynamic light scattering and the seedingactivity was evaluated by RT-QuIC. Histopathology wasused to monitor spongiosis changes, PrPSc accumulationin the brain and astrocytes activation.Results: Spongiosis, PrPSc accumulation in the brainand astrocyte activation were detected in brains from

92 ABSTRACT

90 dpi. However, significant neuronal loss was onlyvalidated at the terminal stage of the disease, ca. 150dpi. Biochemical characterization of brain-derivedPrPSc species at different timepoints is currently inprogress.Conclusions: Our quantitative data measuring NeuNpositive nuclei concur with previous histological assess-ments in indicating that neuronal loss is a late event inprion diseases and thus cannot account for preclinicalreductions in steady-state levels of PrPC. Yet earlierchemical changes can be detected in brain as early as90 dpi. We aim to pinpoint the early PrPSc transitionevents by the FFF technique and understand the causalevents that trigger clinical symptoms of the disease.

References

[1] Sandberg et al., Nature 2011;470:540–542.[2] Mays et al., J Clin Invest. 2014;24(2):847–858.[3] Mays et al., J Virology. 2015;89(24):12,418–1242.[4] Vincenti et al., J Virology. 2016;90(6):3003–3017.[5] Bourgognon et al., Cell Death and Differentiation.

2018;25:1408–1425.[6] Herculano-Houzel, J Neurosci. 2005;5(10):2518–2521.

171. Efficacy of short-term exposure to sodiumhypochlorite (20,000 ppm) or 4% acetic acidsodium lauryl sulfate (4% acetic SDS) to inactivatechronic wasting disease infectivity in a laboratorysetting

Erin E McNulty, Amy V Nalls, Laura A Pulscher,Candace K Mathiason

Colorado State University, Fort Collins, Colorado, USA

CONTACT Erin E McNulty [email protected]

ABSTRACT

Prions are highly resistant to inactivation by conven-tional sterilization processes and disinfecting agents.The Center for Disease Control (CDC) recommenda-tion for decontamination of the infectious agent asso-ciated with prions include exposure to 20,000 parts permillion (ppm) sodium hypochlorite for 30–60 min, orhigh pressure-autoclave at 134 C for 1 h. High pres-sure-high temperature autoclaving is typically used toinactivate prions present on plastics, personal protec-tive equipment and within small quantity tissues/homogenates routinely used by laboratory personnel.Laboratory benches, equipment and instruments areroutinely exposed to 20,000 ppm sodium hypochloritegenerated by diluting household bleach 1:2.5. Due tothe corrosive nature of high sodium hypochlorite

concentrations investigators have searched for effectivemeans to inactivate prions that will result in lessdamage to the laboratory setting. To this end 4% aceticacid sodium lauryl sulfate (4% acetic SDS) has beenincorporated into prion laboratory cleaning regimens.In this study we explore the efficacy of short-termexposure to 20,000 ppm hypochlorite or 4% aceticSDS to inactivate prion infectivity in a laboratory set-ting by use of amplification and bio-assays.

To emulate a more typical laboratory scenario, a filmof brain homogenate (10% w/v) from naïve or chronicwasting disease (CWD)-infected deer was applied to alaboratory bench free of prion contamination. Eachfilm was treated by mist of water, 20,000 ppm hypo-chlorite or 4% acetic acid and left for 30–60 s prior tocleanup. A swab was used to swipe the area postcleanup. Fluid retrieved from the swab was assessedfor infectivity in TgCer5037 mouse bioassay.

We found that 0/9 mice inoculated with 20,000 ppmhypochlorite-treated and 5/6 mice inoculated with 4%acetic SDS-treated CWD inoculum demonstrated clin-ical disease consistent with prion infection.Development of terminal clinical disease in mice inocu-lated with 4% acetic SDS was prolonged by 50% ascompared to no treatment controls. PrPSc depositionand amyloid seeding activity was demonstrated byimmunohistochemistry and PMCA in all mice showingclinical disease. Mice inoculated with similarly treatednaïve brain homogenate remained free of prion disease.

A contemporary dilutional bioassay permitted us toestimate the infectivity remaining in mice inoculatedwith treated inoculum that succumbed to prion infec-tion. We demonstrate that a laboratory cleaning regi-men including short-term exposure to 20,000 ppmhypochlorite results in sterilizing inactivation of CWDprion infectivity while 4% acetic SDS decreased CWDburden by 17%. This study provides findings to guidedevelopment of prion laboratory cleaning procedures.

172. Establishment of PrPCWD extraction anddetection methods in the farm soil

Kyung Je Park, Hoo Chang Park, In Soon Roh, Hyo JinKim, Hae-Eun Kang and Hyun Joo Sohn

Foreign Animal Disease Division, Animal and Plant QuarantineAgency, Gimcheon, Gyeongsangbuk-do, Korea

ABSTRACT

Introduction: Transmissible spongiform encephalopa-thy (TSE) is a fatal neurodegenerative disorder, whichis so-called as prion diseases due to the causative agents(PrPSc). TSEs are believed to be due to the template-

PRION 93

directed accumulation of disease-associated prion pro-tein, generally designated PrPSc. Chronic wasting dis-ease (CWD) is the prion disease that is known spreadhorizontally. CWD has confirmed last in Republic ofKorea in 2016 since first outbreak of CWD in 2001.The environmental reservoirs mediate the transmissionof this disease. The significant levels of infectivity havebeen detected in the saliva, urine, and faeces of TSE-infected animals. Soil can serve as a stable reservoir forinfectious prion proteins. We found that PrPCWD canbe extracted and detected in CWD contaminated soilwhich has kept at room temperature until 4 years after0.001 ~ 1% CWD exposure and natural CWD-affectedfarm soil through PBS washing and sPMCAb.Materials and Methods: Procedure of serialPMCAb. CWD contaminated soil which has kept atroom temperature (RT) for 1 ~ 4 year after 0.001%~1% CWD brain homogenates exposure for 4 monthscollected 0.14 g. The soil was collected by the samemethod once of year until 4 year after stop CWDexposure. We had conducted the two steps. There aretwo kinds of 10 times washing step and one amplifi-cation step. The washing step was detached PrPSc

from contaminated soil by strong vortex with max-imum rpm. We harvest supernatant every time by 10times. As the other washing step, the Washed soilwas made by washing 10 times soil using slow rotatorand then harvest resuspended PBS for removing largeimpurity material. Last step was prion amplificationstep for detection of PrPCWD in soil supernatant andthe washed soil by sPMCAb. Normal brain homoge-nate (NBH) was prepared by homogenization ofbrains with glass dounce in 9 volumes of cold PBSwith TritonX-100, 5 mM EDTA, 150 mM NaCl and0.05% Digitonin (sigma) plus Complete mini proteaseinhibitors (Roche) to a final concentration of 5%(w/v) NBHs were centrifuged at 2000 g for 1 min, andsupernatant removed and frozen at −70 C for use.CWD consisted of brain from natural case in Koreaand was prepared as 10%(w/v) homogenate. Positivesample was diluted to a final dilution 1:1000 in NBH,with serial 3:7 dilutions in NBH. Sonication wasperformed with a Misonix 4000 sonicator with ampli-tude set to level 70, generating an average output of160W with two teflon beads during each cycle. Oneround consisted of 56 cycles of 30 s of sonicationfollowed 9 min 30 s of 37°C incubation. WesternBlotting (WB) for PrPSc detection. The samples(20 µL) after each round of amplification weremixed with proteinase K (2 mg/ml) and incubated37°C for 1 h. Samples were separated by SDS-PAGE

and transferred onto PVDF membrane. After block-ing, the membrane was incubated for 1 h with 1stantibody S1 anti rabbit serum (APQA, 1:3000) anddeveloped with enhanced chemiluminescence detec-tion system.Results: We excluded from first to third supernatant inview of sample contamination. It was confirmed abnor-mal PrP amplification in all soil supernatants fromfourth to tenth. From 0.01% to 1% contaminatedwashed soils were identified as abnormal prions.0.001% contaminated washed soil did not show PrPspecific band (Fig 1). The soil was collected by thesame method once of year until 4 year after stopCWD exposure. After sPMCAb, there were noPrPCWD band in from second to fourth year 0.001%washed soil. but It was confirmed that the abnormalprion was amplified in the washing supernatant whichwas not amplified in the washed soil. we have decidedto use soil supernatant for soil testing (Fig. 2). Afterthird rounds of amplification, PrPSc signals observed inthree out of four sites from CWD positive farm play-ground. No signals were observed in all soil samplesfrom four CWD negative farm (Fig. 3).Conclusions: Our studies showed that PrPCWD persistin 0.001% CWD contaminated soil for at least 4 yearand natural CWD-affected farm soil. When cervid rein-troduced into CWD outbreak farm, the strict deconta-mination procedures of the infectious agent should beperformed in the environment of CWD-affected cervidhabitat.

References

[1] Nagooka K et al., Sensitive detection of scrapie prionprotein in soil. Biochem Biophys Res Commun.2010;397:626–630.

[2] Georgsson G et al., Infectious agent of sheep scrapiemay persisit in the environment for at least 16 years. Jof Gen Virol. 2006;87:3737–3740.

[3] Saunders SE et al., Prions adhere to soil minerals andremain infectious. Plos Pathog. 2006;2:296–302.

[4] Tamguney G et al., Asymptomatic deer excerete infec-tious prions faces. Nature. 2009;529–532.

173. Antisense oligonucleotides for the treatmentof neurodegenerative diseases

Hien T Zhao, Holly Kordasiewicz, Dan Norris, AnneSmith, Roger Lane, C. Frank Bennett and Eric Swayze

Ionis Pharmaceuticals, Inc. Carlsbad, CA,USA

CONTACT Hien T Zhao [email protected]

94 ABSTRACT

ABSTRACT

Antisense oligonucleotides (ASOs) are emerging as aviable therapeutic approach to alter production of pro-teins implicated in currently untreatable neurodegen-erative diseases. ASOs are single stranded nucleotides,typically 20 bases in length, that bind complementarytarget RNA through Watson and Crick hybridization.Depending on ASO design, hybridization can lead toselective degradation of the target RNA, alteration ofRNA splicing or another highly specific post-transcrip-tional modification. Foundational science tools areavailable to facilitate transition from the bench to theclinic, as evidenced by regulatory approval of an ASOfor the treatment of spinal muscular atrophy and initia-tion of clinical trials for ASOs in Huntington’s disease,amyotrophic lateral sclerosis and other neurologicalconditions. Tools that guide drug development andmitigate risk include pharmacology studies in rodentsto demonstrate proof-of-concept phenotypic benefit ofASOs and biodistribution studies in larger species tocharacterize pharmacokinetics and pharmacodynamicsof the ASO and key target-engagement markers.Additionally, PKPD models can be built using preclini-cal data to predict ASO effects in humans, enablinginformed selection of ASO doses for clinical trials andinterpretation of data collected in the clinic. Here, wedescribe our experiences in ASO development for neu-rodegenerative diseases, including proof-of-conceptand biodistribution animal studies, PKPD model-build-ing and clinical trial design and interpretation. Theseexperiences provide a blueprint for advancement ofASO therapies designed to treat neurodegenerative dis-eases, including prion disease.

174. Developing a gene reporter system allowingreal-time in-vivo monitoring of prion diseaseprogression

Matthew J. Martina and Stephanie A. Bootha,b

aUniversity of Manitoba; bNational Microbiology Laboratory, PublicHealth Agency of Canada

CONTACT Matthew J. Martin [email protected]

ABSTRACT

Introduction: Prion diseases are a fatal neurodegenerativedisease caused by the misfolding of the cellular prionprotein, PrPC into an infectious isoform, PrPSc.Accumulation of this infectious isoform leads to thedamage and death of neurons, and the progression of the

disease. The molecular events involved in the pathogenesisof this disease are poorly understood and uncovering themhas proven challenging due to the difficulties of identifyingand isolating degenerating neurons for analysis. Ultimately,the aim of this project is to engineer adeno-associatedviruses (AAV) to deliver genetic tools into neurons todrive the expression of reporter molecules during neuro-degeneration. This would enable real-time monitoring ofthe disease in vivo, and identification of specific neurons tobe isolated for further analysis.Materials and Methods: AAV rh10 was used to deliver agenetic construct containing a short hairpin RNA sequencetargeting the cellular prion gene to test the expressionsystem. This virus construct was injected into newbornmice via their superficial temporal vein. This short hairpinRNA is designed to post-transcriptionally silence prionexpression, inducing immunity against prions to the trans-duced cell. The genetic construct also expresses a redfluorescent protein to be used as a biomarker signallingtransduced cells. At 7 weeks of age, the mice were infectedwith RockyMountain Laboratory prions, or mock-infectedas a control. Brain, liver, lungs, spleen, and serum sampleswas collected at 60, 90, and 141 days post-infection. IHC isused to determine the presence of the virus in these tissuesand Laser Capture Microdissection used to specificallyisolate transduced cells for further analysis of their RNAand protein content.Results: The distribution of the AAV rh10 throughoutmultiple regions of mouse brain will be described aswell as the relative transduction of neurons versusmicroglia and astrocytes. In addition, the kinetics ofthe vector concentration in the brain over time will bedescribed. This work is the basis for engineering vectorscapable of targeting specific neurons in mouse braintissue in future studies to understand prion pathogen-esis and prion strains in vivo.

175. Environmental pathways of CWD prions

Alsu Kuznetsovaa, Debbie McKenzieb and Judd M.Aikena

aAgricultural Life and Environmental Sciences Faculty, University ofAlberta, Edmonton, Canada; bFaculty of Science, University ofAlberta, Edmonton, Canada

CONTACT Alsu Kuznetsov [email protected]

ABSTRACT

Soil can serve as a reservoir and route for horizon-tal transmission of chronic wasting disease (CWD)by interaction with the infectious prion protein

PRION 95

(PrPCWD) shed by infected animals. Prion fate inenvironment is determined by structure of ecosys-tems including soil and vegetation types. In borealecosystems, lichen and shrubs cover sandy textured,quartz-illite soils with surface plant litter horizon.In prairie ecosystems, grassy meadows grow onhumus-enriched, loamy or clay montmorillonitesoils. Once shed onto the soil surface, prion persis-tence in the environment primarily depends on thebinding and transportation capacity of soils. Factorsresponsible for prion migration are soil propertiessuch as mineralogical composition, texture, soilorganic matter content and pH. We investigatedPrPCWD migration in soil profiles by comparingtheir passage through pure soil minerals and diversesoils using lab-scale soil columns. PrP-res wasdetected by western blot or protein misfolding cyc-lic amplification in leachates of columns containingquartz, surface plant litter – soil organic horizon(LFH) of boreal Luvisolic and Brunisolic soils, illite,or the mineral horizons of Luvisol and Brunisol.Infectivity in leachates from quartz, illite andLuvisolic columns was confirmed by bioassay.Analysis of the solid phase of the columns con-firmed the migration of PrPCWD to lower layersin the illite, while the PrPCWD in the montmor-illonite column remained close to the column sur-face. While mineralogical composition of soilsdetermines movement of prions, other constituents,such as soil organic matter, microbial population,composition of soil solutions, might affect prioninfectivity. To investigate the impact of humicacids (a major component of soil organic material),we incubated CWD agent with humic acids forvarious lengths of time. Analysis by western blotand bioassay demonstrated that humic acids canreduce PrPres and infectivity over time. Thus, insoils of temperate regions, including theChernozems of Northern America and Cambisolsof Europe, prions would remain at the soil surfacedue to strong binding to montmorillonite, while inboreal and tundra regions soils (Luvisols, Podzols,and Brunisols), most of the prions would be trans-ported through the plant litter LFH, and partiallythrough the upper soil mineral horizon into lowerhorizons. Although these soils also have lower levelsof humic acids, a soil organic matter compoundthat has the capacity to reduce CWD infectivitylevels, soils of the boreal and tundra regions areless favourable to the horizontal transmission ofCWD as these soils make PrPCWD lessbioavailable.

176. Ultrasensitive detection and seedingselectivity of tau aggregates of Alzheimer diseaseand chronic traumatic encephalopathy

Allison Krausa, Eri Saijoa, Michael M. Metricka, KathyNewellb, Christina J. Sigurdsonc, Gianluigi Zanussod,Bernardino Ghettie and Byron Caugheya

aLPVD, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, USA;bUniversity of Kansas School of Medicine, Kansas City, KS, USA;cDepartment of Pathology, UC San Diego, La Jolla, CA, USA;dUniversity of Verona, Verona, Italy; eIndiana University School ofMedicine, Indianapolis, IN, USA

CONTACT Allison Kraus [email protected]

ABSTRACT

In Alzheimer disease (AD) and chronic traumatic ence-phalopathy (CTE) approximately equivalent amounts of 3-repeat (3R) and 4-repeat (4R) tau isoforms accumulate intau filaments in the brain. To investigate the basis for AD3R/4R tau filament propagation and to improve detectionof 3R/4R tau aggregates as potential biomarkers, we haveexploited the growth mechanism of tau filaments todevelop a highly selective and ultrasensitive cell-free tauseed amplification assay optimized for AD (AD real-timequaking induced conversion or AD RT-QuIC). This reac-tion is based on the capacity of AD tau aggregates to seedformation of amyloids made of certain tau fragments.Seeding activity was detected in AD brains (n = 16) atdilutions as extreme as 107–1010-fold but was 102–106-fold less responsive when seeded with brain from mostcases of other types of tauopathy with roughly comparableloads of tau aggregates composed predominantly of either3R or 4R tau isoforms. AD brains had average seedingactivities that were orders of magnitude higher than Pickdisease brains with predominant 3R tau deposits; however,we previously observed the opposite using our prototypicPick- optimized tau RT-QuIC assay. CTE brains (n = 2)had seed concentrations comparable to some of the ADspecimens, and higher than three of four specimens with3R/4R primary age-related tauopathy (i.e., PART). ADseeds shared properties with the neurofibrillary tanglesfound in AD brains in terms of being sarkosyl-insoluble,protease-resistant, and reactive with tau antibodies.

Moreover, the ADRT-QuIC assay detected as little as 16fg of pure synthetic tau fibrils. The distinctive seedingactivity shown by AD and CTE tau filaments in compar-ison to those of other types of tauopathies suggests atemplating specificity that may underpin their consistentstrain- like propagation as conformers in patients with 3R/4R tau diseases. Importantly, AD RT-QuIC also provides ameans of rapid and ultrasensitive quantitation of 3R/4R tauseeding activity as a biomarker. We are currently adaptingthe assay to the detection of AD and CTE tau seeds in

96 ABSTRACT

biologically accessible specimens to aid in diagnostics andtherapeutics development.

KEYWORDS: Alzheimer disease; chronic traumaticencephalopathy; tau; seed; diagnostics; biomarkers;RT-QuIC

Reference

[1] Kraus A et al., Acta Neuropath. 2018:[Epub ahead ofprint].

177. Assessing the importance of Taumislocalization to the dendrites in Alzheimer’sdisease

Emily A. McNamara, Sue-Ann Mok

Department of Biochemistry, University of Alberta, Edmonton,Canada

CONTACT Emily A. McNamara [email protected]

ABSTRACT

Alzheimer’s disease is a fatal neurodegenerative condi-tion primarily characterized by a decline in cognitivefunction and a loss of motor skills1. This disease affectscells of the central nervous system (neurons) and is theleading cause of all dementias1. While the exact cause ofAlzheimer’s disease is not yet fully understood, one ofthe main components thought to be associated withdisease pathology is the abnormal accumulation of theprotein, Tau2. Tau is a microtubule-associated proteinfound primarily in the neuronal axon, where it plays arole in assembling microtubules that are critical foraxonal transport3. Studies have shown that Tau mislo-calization to the dendritic spines can occur during orpreceding a disease state, causing synaptic disruptionand overall brain dysfunction4. Specific signals in the 3ʹUntranslated Region (UTR) of Tau mRNA transcriptshave been shown to play a key role in subcellular mRNAlocalization and therefore local protein translation5.

Our work aims to modify the human Tau 3ʹ UTR inorder to localize it to the neuronal axon or dendriticspines. Following this, we will be able to explore Tau’sinteractions with various cellular components at eithersubcellular location and potentially uncover key com-ponents involved in disease pathology.

We have designed constructs of the Tau 0N4R isoformwith a modified 3ʹ UTR. The 3ʹ UTR’s utilized are derivedfrom human Tau ormouse Tau, which localize to the axon,or another microtubule associated protein, MAP2, whichlocalizes to the dendrites. While it has been shown thatmouse Tau 3ʹ UTR is capable of directing Tau to the

axons5, this has not yet been replicated with human Tau.The constructs will be transfected into human neuronalcells using lentiviral vectors. Fluorescent antibodies andmRNA probes recognizing specific regions of exogen-ously-expressed Tau will be used to detect the correctlocalization of the protein and transcript. While the analy-sis of Tau interactions at both the axon and dendrites arepart of my long-term project, HEK cell lines that expressTau 0N4R have been transfected with DNAJ2 and Hsc70;two chaperones known to be upregulated duringAlzheimer’s Disease. HEK cells will be induced to expressTau, and potential aggregation will be monitored usingconfocal microscopy. Results from this assay may pinpointDNAJ2 and Hsc70 as key players in Tau aggregation, andresults will be replicated in a human neuronal model.

Understanding the importance of Tau localization inneurodegenerative disease pathology may uncoverpotential key therapeutic targets for the treatment ofAlzheimer’s disease.

KEYWORDS: Alzheimer’s disease; Tau; neurodegen-eration; 3ʹ UTR

References

[1] Alzheimer’s Association. 2015 Alzheimer’s Diseasefacts and figures. Alzheimer’s & Dementia. 2015;11(2015):332–384.

[2] Musi N., Valentine JM. et al., Tau protein aggregationis associated with cellular senescence in the brain.Wiley Aging Cell. 2018:1–13. DOI:10.1111/acel.12,840

[3] Buée L. et al., Tau protein isofroms, phosphorylationand role in neurodegenerative disorders. BrainResearch Reviews. 2000;33:95–130.

[4] Hoover BR. et al., Tau mislocalization to dendriticspines mediates synaptic dysfunction independently ofneurodegeneration. Cell Press Neuron. 2010;68:1067–1081.

[5] Aronov S. et al., Axonal Tau mRNA localization coin-cides with tau protein in living neuronal cells anddepends on axonal targeting signal. The Journal ofNeuroscience. 2001;21(17):6577–6587.

178. Distinct strain of Amyloid beta andpathogenic tau protein in iatrogenic Creutzfeldt-Jakob disease

Jiri G. Safara, Chae Kima, Tracy Haldimana, AnushrutiAshoka, Ignazio Calia, Mark Cohena, Stéphane Haїkb,Piero Parchic, Giorgio Giacconed, Steven Collinse,Nicolas Privatb, Véronique Sazdovitchb, CharlesDuyckaertsb, Diane Kofskeya, Han Wanga, CatrionaMcLeane, Jean-Philippe Brandelb, TetsuyukiKitamotof, Ermias Belayg, Ryan Maddoxh, FabrizioTagliavinid, Maurizio Pocchiari, Brian Applebya,

PRION 97

Pierluigi Gambettij, Ele Gelpik, Ann McKeel, JamesIronsidem and Lawrence Schonbergern

aCase Western Reserve University; bSorbonne Universités; cUniversity ofBologna; dIstituto Neurologico Carlo Besta; eUniversity of Melbourne;fTohoku University; gCenter for Disease Control and Prevention; hCDC;iIstituto Superiore di Sanità; jDepartment of Pathology, Case WesternReserve University, School of Medicine, Cleveland, OH, USA; kHospitalClínic Institut d’Investigacions Biomèdiques August Pi i Sunyer; lBostonUniversity; mUniversity of Edinburgh; nUS Centre for Disease Control andPrevention

ABSTRACT

Objectives: Whether some strains of prion-like proteinaggregates (prionoids) such as amyloid beta (Aβ) andtau propagating in cell culture and transgenic experi-ments can be infectious and transmit the neurodegen-erative diseases from man-to-man is unknown. Wereported recently an increased frequency of amyloid β(Aβ) plaques and angiopathy in a cohort of 27 iatro-genic cases of Creutzfeldt-Jakob disease (iCJD),acquired from the prion-contaminated growth hor-mone (GH) extracted from human cadaveric pituitaryglands, or from cadaveric dura mater grafts (DM) dur-ing neurosurgery. As in human prion diseases, criticalquestions raised by these findings are (a) whether thepathology was transmitted by a unique strain of Aβ ortau with a high potential to initiate or accelerateAlzheimer’s disease (AD), and (b) if such Aβ or taustrains can be identified whithin the spectrum of AD.

Methods: With new biophysical and structural tools,we analysed conformational strain characteristics of Aβand pathogenic tau aggregates in 14 cases of iCJD, andcompared the data with 21 cases of age-matched spora-dic CJD (sCJD), 30 cases of Alzheimer’s diseases withrapid progression (rpAD), 18 cases with slow AD pro-gression (spAD), 7 cases of chronic traumatic encepha-lopathy (CTE), and 8 cases of frontotemporal lobardegeneration (FTLD).Results: The levels of oligomeric and aggregated Aβ42as well as the Aβ42/Aβ40 ratios were in iCJD caseselevated signficantly above the levels found in sCJDcontrols. The conformational characteristics of Aβ42acccumulating in iCJD were distinctly different fromsCJD and reminiscent of small oligomers of Aβ42found most frequently in AD with rapid progression.The levels of detergent-insoluble pathogenic tau aggre-gates were increased in ~14% of iCJD above the levelsfound in sCJD but were generally lower than in AD andother tauopathies. Notably, the conformations of deter-gent-insoluble tau found in hippocampus of iCJD caseswere significantly different from those found in sCJD.Conclusions: The levels of oligomeric and aggregatedforms of Ab42 and pathogenic aggregates of tau proteinare in iCJD cases significantly higher than in sCJD

controls and their conformational characteristics arein the range corresponding to the strains found mostfrequently in rapidly progressive AD. Although thecontributions of other factors, including GH deficiencycannot be discounted, our findings argue that this ADpathology was acquired by a distinct strain of amyloidbeta conformers through a mechanism resembling thatof prion diseases.

179. PrPCWD detection in CWD-infected TgElk micemodel using RT-QUIC

Hyun Joo Sohn, Kyung Je Park, In Soon Roh, Hyo JinKim, Hae Eun Kang

Foreign animal disease division, Animal and Plant QuarantineAgency, Gimcheon, Gyeongsangbuk-do, Korea

ABSTRACT

Introduction: Chronic wasting disease (CWD) is the onlyprion disease affecting free-ranging animals, reported inNorth America, South Korea, and Norway. Unlike inmost other prion disease CWD agents are shed in blood,urine, and faeces which most likely contribute to thehorizontal transmission between cervid species. Thedevelopments of amplification-based seeding assays havebeen instrumental in the detection of low levels of prionsin clinical samples. Using real-time quaking-induced con-version (RT-QUIC), we established an ultrasensitivedetection method for PrPCWD in the urine from CWD-infected sequentially sampled transgenic mice overex-pressing elk prion protein (TgElk mice). In addtion, RT-QUIC was performed in the kidney and brain of thesesmice model to trace abnormal prion.Materials and Methods: 44 brain and kidney, urinesamples from sequentially collected from CWD-infected TgElk mice (TgElk CWD) were stored at−80°C. In brain and kidney, 10% (w/v) homogenatewas prepared in 0.9% sterilized saline. In urine 100 µLof each sample was mixed with 10 µL 2.8% sodiumphosphotungustic acid (NaPTA) and incubated for1hr at 37°C with shaking at 1,350 rpm. Samples werecentrifuged for 30 min at 16,100 g. The pellet wasresuspended in 10 µL of 0.1% SDS/PBS for 30 min at55°C. RT-QUIC reactions were set up in 96-well clearbottom optic plates and consisted of 98 µL RT-QUICbuffer [final concentrations of 1XPBS, 1 mM EDTA,10 µM Thioflavin, 300 mM NaCl buffer and 0.1 mg/mlrecombinant Syrian hamster recombinant protein (23–231), and 2 µL of sample. The RT-QUIC assay wasperformed on a FLUOstar Omega fluorescence platereader that was preheated to 42°C for 60 h with 90 sshaking at 700 rpm followed by 1 min incubation.

98 ABSTRACT

Results: Five randomly selected mice were sequentiallyculled on every 15 days from 30dpi to 120dpi duringCWD infected TgElk mice reached terminal stage.Rough hair coats among clinical signs were showedfrom 90 dpi. PrPCWD in the brain in TgElk CWD wasdetectable persistently from early stages (30dpi), and inthe kidney PrPCWD was also detectable in clinical andterminal stages (90 dpi and 120dpi). PrPCWD in theurine in TgElk CWD reached the highest levels at120dpi. NaPTA/RT-QUIC was applied to measurePrPCWD in urine samples collected on every 15 daysfrom 30dpi to 120dpi when CWD infected TgElk micereached terminal stage. PrPCWD in the urine in TgElkCWD reached the highest levels at 90dpi. PrPCWD wasalso detectable in late and terminal stages (120dpi).Conclusions: We demonstrate that CWD prions can bedetected by RT-QUIC or NaPTA/RT-QUIC in thebrain, kidney and urine of TgElk mice at the earlyand terminal stages of disease. Based on these data,we suggest that PrPCWD is excreted into only urineuntil 90 dpi and then slowly accumulated in kidney.Our results can be used in designing future study ofCWD pathogenesis in TgElk mice.

References

[1] Henderson DM et al., Rapid antemortem detection ofCWD prions in deer saliva. PLOS one. 2013:e74377 1–12

[2] Nagooka K, Yoshika M, Shimozaki N. et al., Sensitivedetection of scrapie prion protein in soil. BiochemBiophys Res Commun. 2010;397:626–630.

180. Cell assay detection of chronic wastingdisease (CWD) prion infectivity

Su Bi Ahna, Hyo-Jin Kima, Jifeng Bianb, In-Soon Roha,Hae-Eun Kanga, Hyun-Joo Sohna

aForeign Animal Disease Division, Animal and Plant QuarantineAgency, Gimcheon, Gyeongsangbukdo, Korea; bPrion ResearchCenter (PRC), Colorado State University, Fort Collins, CO, USA

CONTACT Hyun-Joo Sohn

ABSTRACT

Chronic wasting disease (CWD) is a prion diseaseaffecting the animals in family Cervidae. Prions arethe infectious agents that cause prion disease. Mouseprion bioassay is the most commonly used method fordetermine prion infectivity. Application of more effi-cient measurement method was required as severalcases of CWD have been reported in South Korea. Inthis study, we adapted the modified cervid prion cellassay (CPCA) using Elk21− cells. The susceptible

Elk21− cells were exposed to various CWD brainhomogenates, ranging from 10−2 to 10−5. The CPCAtiters of elk CWD brain homogenates were 105.8 [2001(E190Y+229Y)] and 106.5 [16FC050] units/g of brain,respectively. We also found the CWD prion titer of redand sika deer brain inocula in 2016 CWD cases, produ-cing CPCA titers of 105.8 and 105.4 units/g of brain.These results were confirmed by western blotting.Comparing mouse titers of mouse ic LD50/g of cervidbrain with CPCA titers, sensitivity of the cell-basedmeasurement was similar to that of the mouse bioassay.Also, CPCA titers does not depend on CWD species,relate with mouse bioassay survival time. Consequently,our results demonstrate that CWD prion infectivity canbe efficiently and quantitatively detected by cell basedtitration of CWD.

181. Application of the TSE-infected cell and RT-QUIC assay to discover novel natural products

Hyo-Jin Kim, Su Bi Ahn, In-Soon Roh, Kyung-Je Park,Hae-Eun Kang, Hyun-Joo Sohn

Foreign Animal Disease Division, Animal and Plant QuarantineAgency, Gimcheon, Gyeongsangbukdo, Korea

CONTACT Hyun-Joo Sohn

ABSTRACT

Conformational conversion of the normal prion pro-tein (PrPC) into an infectious PrPSc causes pathogenesisin prion diseases or transmissible spongiform encepha-lopathies (TSEs). TSE-infected cell lines have been effi-ciently used to study this conversion process, as well asto screen for potentially effective anti-prion com-pounds. We employed fragment molecular orbital(FMO)-based virtual screening and BSE-infected cell-based assay system to discover two natural productswith antiprion activity exhibited good binding interac-tions, with hotspot residues within the PrPC bindingsite, and effectively reduced PrPSc levels in a standardscrapie cell assay (SSCA). We also investigated theprion inhibition ability of two novel natural productsin Elk 21+ (CWD) or SMB (Scrapie) cells. Cytotoxicitytests showed no significant cell death in either BNP-03or BNP-08 treatment. Elk21+ and SMB cells were sub-jected to two effective natural products for reducingPrPSc, BNP-03, and BNP-08. Efficiency of PrPSc clear-ance for two natural products was estimated by evalu-ating the signal sensitivity for PrPSc on SSCA, WB, andEIA. Antiprion activity of the compounds was detectedby RT-QUIC assay seeded with TgElk CWD (E190Y+229Y). These natural products showed the inhibitoryeffects in the TSE-infected cells. Further experiments in

PRION 99

animal models will be necessary to confirm the effect oftwo novel natural products in vivo.

182. Species and sex differences in contact duringthe breeding season may explain variation inChronic Wasting Disease prevalence

Kelsey Saboraki and Susan Lingle

Department of Biology, University of Winnipeg

CONTACT Kelsey Saboraki [email protected]

ABSTRACT

Background: Chronic Wasting Disease (CWD) is atransmissible and fatal neurodegenerative prion dis-ease that threatens wild populations of ungulates.Prevalence of CWD is higher in mule deer than inwhite-tailed deer and in males than in females ofboth species. Prions are spread through direct andindirect contact with saliva, urine, faeces and to anunknown extent, glandular secretions. Species andsex differences in rates of direct contact with infectedindividuals or indirect contact with environmentalcontaminants may result in species and sex differ-ences in pathogen transmission and disease preva-lence. Social behaviours and the potential forpathogen transmission change seasonally, and inter-actions during the breeding season may explain dif-ferences in prevalence. We therefore tested thehypothesis that species and sex differences in beha-viour during the breeding season underlie speciesand sex differences in CWD prevalence in mule andwhite-tailed deer.Methods: We observed behavioural interactionsbetween deer during the breeding season on an opengrassland site, the McIntyre Ranch, in southern Albertato assess whether species and sex differences in directphysical contact (deer to deer) and indirect contact(deer to environment to deer) correspond to the higherprevalence of CWD in mule deer and in males. Weobserved 86 deer (21 mule deer females, 21 mule deermales, 20 white-tailed females, and 24 white-tailedmales) and recorded the occurrence and duration ofdeer-deer and deer-environment-deer contact and thenumber of partners with which deer engaged in contactwith over time.Results and Discussion: We found that males exhi-bit sex-specific behaviours, such as marking vegeta-tion and flehmen, that have the potential to elevatetheir risk of prion transmission relative to females.During courtship interactions, between sex contactwas more common in mule deer than in white-tailed

deer, which could explain species differences in pre-valence. As females are in estrous for only 1–2 dayswhile a successful male engages in high-risk court-ship throughout the rut, these results show thatmales of both species should have a higher cumula-tive potential for prion transmission throughout thebreeding season. By collecting empirical data on thefine-scale behaviour of sympatric mule deer andwhite-tailed deer during the breeding season, weidentified mechanisms that may be responsible forthe higher prevalence of CWD in mule deer and inmales.

KEYWORDS: Chronic wasting disease; mule deer;white-tailed deer; behaviour; transmission

183. Morphometric analysis of PrP deposition intwo models of prion diseases

Pawel P. Liberski a, Agata Gajos b, Beata Sikorska a

and Janusz Moryś c

aLaboratory of Electron Microscopy and Neuropathology,Department of Molecular Pathology and Neuropathology, Chair ofOncology, Medical University Lodz, Poland; bDepartment ofExtrapyramidal Diseases, Chair of Rehabilitation, MedicalUniversity of Lodz, Lodz, Poland; cDepartment of Anatomy andNeurobiology, Medical University of Gdansk, Gdansk, Poland

CONTACT Pawel P. Liberski

ABSTRACT

The Echigo-1 strain of Creutzfeldt-Jakob disease (CJD)was isolated from a case of a 33-years-old female with apanencephalopathic type of CJD. The Fujisaki(Fukuoka 1) was isolated from a case of Gerstmann-Straussler-Scheinker disease (GSS)Material andMethods: Themorphometrical analysis wasdone using the Zen Blue v. 2.6 and its automated analysismodule for calculation of percentage of total area of allimmunopositive objects in relation to the area of themeasurement frame as well as the mean intensity of greyvalues of an analysed region and the pathological changes.Results: The significant differences in the distribution ofpathological changes were observed in the studied twogroups of animals. In the Echigo-1 animals the mostintense changes were observed in the lateral dorsal nucleus(49.8% of studied area was occupied by the pathologicalprotein) and ventrolateral nucleus (20.3%) of the thalamus,auditory (37.8%) and somatosensory (32.9%) cortices andthe hilus (22.4%), and CA2 (28.6%) sector of the hippo-campus. The smallest changes were present in the dentategyrus (1.4%) and putamen (5.1%). The highest reaction ofthe microglia was observed in the somatosensory cortex,lateral dorsal nucleus, and in the hippocampal sectors. Very

100 ABSTRACT

weak reaction of the microglia was present in the dentatehilus, putamen, and ventrolateral thalamus.

In the second group of animals the most dramaticchanges were observed in the thalamus (ventrolateralnucleus – 56.2%; lateral dorsal nucleus – 50.3%, andlateral posterior nucleus – 40.7%) as well as in thehippocampus (CA1 – 48.1%, CA3 – 40.3%, and CA2– 35.6). The moderate reaction was present the soma-tosensory and auditory cortex, basolateral amygdala,and putamen. The weak reaction was observed in thedentate gyrus and endopiriform nucleus. The intensivereaction of microglia was present in the somatosensorycortex, in the thalamus and basolateral amygdala. Thesmall reactivity of the microglia was observed in thedentate gyrus and its hilus as well as in the CA1 sector.The microglia reactivity was almost twice bigger in thepresent in the animals injected by the Fujisaki strainthan Echigo-1, also the intensity of immunoreactivityfor prion protein distribution was biggest in the firstgroup of animals.

Funding

PPL and BS are supported by National Science CentrePoland, grant number UMO-2015/19/B/NZ4/03234

184. Complex pathways for native folding andmisfolding of SOD1 observed directly by single-molecule force spectroscopy

Supratik Sen Mojumdar, Craig R. Garen and Michael T.Woodside

University of Alberta

CONTACT Michael T. Woodside

ABSTRACT

Superoxide dismutase 1 (SOD1) is a β-barrel dimerthat undergoes prion-like misfolding in the contextof ALS, but its misfolding mechanism remainsunclear. We studied the folding of SOD1 monomersand dimers by using laser tweezers to unfold andrefold single molecules held under mechanical ten-sion. Monomers were found to fold via numerousintermediate states, in contrast to the two-state fold-ing deduced from ensemble studies. A novel pathwayanalysis indicated that these intermediates involve theaddition of individual β-strands to the native struc-ture. Several misfolded states were also observed,branching off from the native pathway after the for-mation of a stable native-like core [Sen Mojumdar etal., Nat. Commun. 2017]. In dimers, the interfacebetween domains changed the folding noticeably, in

several ways. First, the number of intermediates waslower than expected for independent monomers,indicating increased cooperativity. Second, thedimer interface was found to modulate the foldingpathways by changing the relative stability of themonomer segments close to the interface. Third, thedimer misfolded only half as much as expected fromthe monomer misfolding rate, suggesting the inter-face helps protect against misfolding, in part byreducing the prevalence of partially-folded intermedi-ates that lead to misfolding. Finally, inserting thepoint mutation G85R, which is associated with famil-ial ALS, vastly increased the rate of misfolding.

185. Probing cortico-cortical and hippocampal-cortical interactions in a second-generation mousemodel of Alzheimer’s disease

Surjeet Singh, Jogender Mehla, Robert J. Sutherlandand Majid Mohajerani

Department of Neuroscience, Canadian Center for BehavioralNeuroscience, University of Lethbridge, Alberta, Canada

CONTACT Surjeet Singh

ABSTRACT

It is known that Alzheimer’s disease (AD) is associatedwith defects of synaptic connectivity. Such defects maynot be restricted to local neuronal interactions but mayextend to long-range brain activities, such as slow-waveoscillations that are particularly prominent duringnon–rapid eye movement (non-REM) sleep and areimportant for integration of information across distantbrain regions involved in memory consolidation. Usinga newly characterized knock-in mouse model of AD(n = 8, 12 months old) that has humanized amyloidbeta (Aß) sequence knocked-in to the murine amyloidprecursor protein gene along with the Swedish, Arctic,and Beyreuther/Iberian mutations (APP NL-G-F), andwide-field optical imaging of cortical voltage responses(VSDI) combined with local field potential (LFP)recording from CA1, we investigated that how Aßdeposition in these mouse model impacts cortical func-tional connectivity and how different cortical regionsinteract with hippocampus during slow wave sleep. Wefound that Aß deposition in knock-in mouse model ofAD (APP NL-G-F) impairs cortical functional connec-tivity and intrinsic hippocampal circuit that generatesSWR. Coordination of cortical up-states and hippo-campal sharp-wave ripples (HPC-SWR) during slowwave sleep is believed to play a major role in theconsolidation of recently acquired memories. It wasobserved that HPC-SWR show strong correlation with

PRION 101

cortical activation in retrosplenial cortex (RSC).However, the magnitude of ripple power and corticalactivation in RSC is reduced in AD mice as comparedlittermate controls, suggesting impairment in hippo-campal-cortical interaction during slow wave sleep.We further analysed cortical VSDI data form 19 regionof interests (ROIs) with two nonlinear methods:entropy (En) and auto mutual information (AMI). Enquantifies regularity in data, while AMI detects linearand nonlinear dependencies in time series. Weobserved that En was lower in AD mice, and AMIdecreased more slowly with time delays in AD as com-pared to control mice, for both En and AMI significantdifferences were observed in occipital, somatosensoryand motor cortex (p <0.05). These results provide newinsights into brain dysfunction associated with Aßdeposition in APP NL-G-F mouse model of AD.

186. Serial detection of hematogenous prions inCWD-infected deer

Amy V. Nalls, Erin E. McNulty, Nathaniel D. Denkers,Edward A. Hoover and Candace K. Mathiason

Department of Microbiology, Immunology, and Pathology,Colorado State University, Fort Collins, CO, USA

CONTACT Amy V. Nalls [email protected]

ABSTRACT

Blood contains the infectious agent associated withprion disease affecting several mammalian species,including humans, cervids, sheep, and cattle. It hasbeen confirmed that sufficient prion agent is presentin the blood of both symptomatic and asymptomaticcarriers to initiate the amyloid templating and accumu-lation process that results in this fatal neurodegenera-tive disease. Yet, to date, the ability to detect blood-borne prions by in vitro methods remains difficult.

We have capitalized on blood samples collected fromlongitudinal chronic wasting disease (CWD) studies inthe native white-tailed deer host to examine hemato-genous prion load in blood collected minutes, days,weeks and months post exposure. Our work hasfocused on refinement of the amplification methodsRT-QuIC and PMCA. We demonstrate enhanced invitro detection of amyloid seeding activity (prions) inblood cell fractions harvested from deer orally-exposedto 300 ng CWD positive brain or saliva.

These findings permit assessment of the role hema-togenous prions play in the pathogenesis of CWD andprovide tools to assess the same for prion diseases ofother mammalian species.

187. Impact of new Creutzfeldt-Jakob diseasemouse models in prion research

Juan Maria Torresa, Juan Carlos Espinosaa, OlivierAndreolettib, Isidre Ferrerc, Inga Zerrd, FrancLlorensc,d

aCentro de Investigación en Sanidad Animal-INIA, Madrid, Spain;bFrench National Institute for Agricultural Research (INRA)-ENVTUMR 1225, Toulouse, France; cBellvitge Biomedical ResearchInstitute, Barcelona, Spain and Network Center for BiomedicalResearch of Neurodegenerative Diseases, Institute Carlos III,Ministry of Health, Spain; dDepartment of Neurology, UniversityMedical School, Göttingen, Germany

CONTACT Franc Llorens [email protected]

ABSTRACT

Studying animal prion diseases models is an appealingapproach to dissect the temporal-dependent physio-pathology of human prion diseases. In the recentyears, new mouse models have been developed basedon the genetic deletion of the endogenous mouse prionprotein gene (PrnP 0/0) and the overexpression of thehuman prion protein (PRNP). After their inoculationwith human brain homogenates, the animals develop aprion pathology, which highly recapitulates the neuro-pathological and biochemical features of human priondiseases in a subtype-dependent manner.

These humanized mouse models have shown to becrucial for the understanding of the genetic susceptibilityto prions, the transmissibility properties of different prionsubtypes and the zoonotic potential of prion strains.However, the precise molecular and signalling featuresoccurring during the progression of disease pathology inthese models have not been yet scrutinized in detail.

We used the tg340-MMmousemodel (transgenicmiceoverexpressing the PRNP with Met/Met at its codon 129inoculated with sCJD MM1 brain homogenate) as amodel of sCJD subtype MM1 in order to gain insightinto the molecular mechanism driving to human prionpathogenesis. Targeted and screening studies were carriedout in a temporal and regional-dependent fashion andmain outcomes were systematically compared to thoseobtained in sCJD MM1 post-mortem cases.

In the tg340-MMmice, we detected temporal and regio-nal dependent alterations in neuro-inflammatory profiles,miRNA, gene expression signatures, prion protein deposi-tion patterns and pathogenic-associated cell signallingpathways that were highly similar than those detected insCJDMM1 cases. Additionally, the study of the tg340-MMmice allowed us to profile the molecular signatures at pre-clinical and early-clinical stages of the disease.

Altogether, we proved the usefulness of new trans-genic humanized CJD models as a valid tool for the

102 ABSTRACT

study of the molecular features associated with humanprion diseases.

188. Full atomistic model of PrPSc structure andconversion

Giovanni Spagnollia, Marta Rigolia, Simone Oriolia,b,Alejandro M Sevillanoc, Pietro Facciolia,b, HolgerWilled, Emiliano Biasinia, Jesus R Requenae

aUniversity of Trento, Italy; bItalian Institute for Nuclear Physics(INFN) - TIFPA, Italy; cUniversity of California San Diego, USA;dUniversity of Alberta, Canada; eUniversity of Santiago deCompostela, Spain

CONTACT Giovanni Spagnolli [email protected]

ABSTRACT

Since their discovery, prions have stood as enigmaticagents that defy the classical concept of genetic inheri-tance due to the capacity of propagating their conforma-tionally encoded information in absence of nucleic acid[1].However, after more than 30 years, the high resolutionstructure of PrPSc, required to understand the molecularfeatures of prion infectivity, has remained elusive [2].Different PrPSc models have been proposed but they arenow inconsistent with recent results [2].In this work,using molecular modelling techniques, we constructedan all-atom model of mouse PrPSc, derived from theintegration of a wide array of experimental constraints,such as: (i) cryo-EM and X-ray fiber-diffraction studies,which showed that the fold of a mouse infectious PrP iscompatible with a 4-rung-β-solenoid (4RβS) architecture[3,4];(ii) circular dichroism and FTIR spectroscopy,which ruled out the presence of α-helices [5];(iii) massspectrometry analyses, indicating the presence of an intactdisulphide bond between residues C178 and C213 [6],aswell as mapping Proteinase K sensitive residues, whichreflect amino acids likely excluded from the resistant coreof the protein [7];and (iv) the possibility of accommodat-ing complex glycans at positions N180 and N196 [8]. Thestability of this new PrPSc model, assessed by MolecularDynamics simulations, was found to be comparable tothat of the prion forming domain of Het-s, a naturally-occurring β-solenoid protein. Finally, we coupled theinformation of the 4RβS structure with Ratchet andPawl Molecular Dynamics [9],to perform for the firsttime a simulation of the conformational transition fromPrPC to PrPSc. The result is a transition pathway withatomistic resolution in which the C-terminal rung of thesolenoid acts as a primary conversion surface for PrPCunstructured N-terminus, followed by a cascade of con-formational changes where each newly formed rung tem-plates the formation of the following one, ultimately

leading to the complete conversion of PrPC into PrPSc.This structure is, to the best of our knowledge, the firstphysically plausible PrPSc model, and represents a uniqueworkbench for interpreting future structural data or avail-able biological evidence, such as the effects of variations inthe PRNP gene favoring or disfavoring prion propagation.Furthermore, it allowed us to reconstruct how the infor-mation encoded into the conformation of a protein couldbe propagated in a directional fashion: a concept under-lying the infectious nature of prions.

KEYWORDS: PrPSc Structure; Prion Propagation;Molecular Modelling; Molecular Dynamics

References

[1] Prusiner SB. Science. 1982;216(4542):136-144.[2] Wille H, Requena JR. Pathogens. 2018;7(1):20.[3] Vazquez Fernandez E, et al. PLoS Pathog. 2016;12(9):

e1005835.[4] Wille H, et al. Proc Natl Acad Sci USA. 2009;106

(40):16990-16995.[5] Requena JR, Wille H. Prion. 2014;8(1):60-66.[6] Welker E, et al. J Biol Chem. 2002;277(36):33477-

33481.[7] Vazquez Fernandez E, et al. PLoS Pathog. 2012;14(1):

e1006797.[8] Baskakov IV, Katorcha E. Front Neurosci. 2016;10:358.[9] Tiana G, Camilloni C. J Chem Phys. 2012;137

(23):235101.

189. Prion substrain specific tropism for thespleen: role of PrPC expression levels

Fabienne Reinea, Laetitia Herzoga, Philippe Tixadora,Thanh-Lan Laïa, Johann Castilleb, Bruno Passetb,Human Rezaeia, Olivier Andréolettic, Jean-LucVilotteb, Hubert Laudea and Vincent Béringuea

aVIM, INRA, Université Paris-Saclay, Jouy-en-Josas, France; bGABI,INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France;cIHAP, INRA, ENVT, Toulouse, France

CONTACT Vincent Béringue [email protected]

ABSTRACT

Prions are proteinaceous pathogens formed from mis-folded conformers (PrPSc) of the host encoded cellularprion protein (PrPC). Prions replicate by templating theconversion and polymerization of PrPC by an autocata-lytic process. Multiple strains of prions are recognizedphenotypically within the same host species. Strains areconformational variants of PrPSc at the level of the ter-tiary and the quaternary structure. In infected hosts, prionstrains exhibit a stereotyped biological phenotype, thatcan include specific tropism for the lymphoid tissue.

PRION 103

Prion isolates and strains can be composed of severalsubstrain components. These substrains can propagatein distinct tissues of the same infected individual (e.g.brain and spleen), in both homotypic and heterotypicPrP transmission contexts [1–3]. The reason for suchtissue selectivity is unclear. Previously, we demonstratedthat PrPC expression levels in the brain govern prionsubstrain selection during homotypic PrP transmissionevents [4]. Thus, transmission of natural sheep scrapieisolates (termed LAN) to ovine PrP transgenic miceexpressing different levels of PrPC in the brain resultedin the phenotypic expression of the dominant LAN sub-strain in mice expressing near physiological PrPC levels,whereas a minor substrain was preferentially selected onhigh-expressors.

Considering that PrPC expression levels are markedlydecreased in the spleen compared to the brain in wild-typeand PrP transgenic mouse models1, we questionedwhether PrPC amount could drive prion substrain spleen-selectivity. The outcome of LAN sheep scrapie isolatestransmission in the spleen from high-expressors correlatedwith the replication rate dependency on PrPC amount,with exclusive and prominent colonization of the spleenby the substrain preferentially replicating on low-expres-sors. After intraperitoneal inoculation of LAN isolates,such early spleen colonization allowed the neuropatholo-gical expression of this otherwise brain-unfavoured sub-strain. In addition, a pair of strain variants resulting fromthe adaptation of cortical MM2CJD subtype to ovine high-expressors, and exhibiting differing brain vs. spleen trop-ism in these mice [3], showed opposite tropism in low-expressors.

Collectively, our results support the view that PrPCexpression levels are instrumental in prion substrainspecific tropism for the lymphoid tissue.

References

[1] Beringue V. et al., Science. 2012;335:472–475.[2] Beringue V. et al., PLoS One. 2008;3:e1419.[3] Chapuis J. et al., Acta Neuropathol Commun.

2016;4:2–15.[4] Le Dur A. et al., Nat Commun. 2017;8:14,170.

190. Independent amplification of co-infected longincubation period low conversion efficiency prionstrains

Thomas E. Eckland, Ronald A. Shikiya and Jason C.Bartz

Department of Medical Microbiology and Immunology, School ofMedicine, Creighton University, Omaha, NE, USA

CONTACT Jason C. Bartz [email protected]

ABSTRACT

Prion diseases are caused by a misfolded isoform of theprion protein, PrPSc. Prion strains are encoded by strain-specific conformations of PrPSc and prions can interferewith each other when a long-incubation period straininhibits the conversion of a short-incubation periodstrain. Prion strain interference influences the emer-gence of a strain from a mixture and prion straindynamics. It is unknown, however, if two long-incuba-tion period strains can interfere with each other. Here,we show that co-infection of animals with combinationsof long-incubation period prion strains failed to identifyevidence of strain interference. To exclude the possibilitythat this inability of strains to interfere in vivowas due toa failure to infect common populations of neurons weused protein misfolding cyclic amplification strain inter-ference (PMCAsi). Consistent with the animal bioassaystudies, PMCAsi indicated that both co-infecting strainswere amplifying independently, suggesting that the lackof strain interference is not due to a failure to target thesame cells but is an inherent property of the strainsinvolved. Importantly PMCA reactions seeded withlong incubation-period strains contained relativelyhigher levels of remaining PrPC compared to reactionsseeded with a short-incubation period strain.Mechanistically we hypothesize that co-infection withprion strains with relatively low prion conversion effi-ciencies, the abundance of PrPC is not limiting in vivo orin vitro. Overall, this observation changes the paradigmof the interactions of prion strains and has implicationsfor interspecies transmission and emergence of prionstrains from a mixture.

191. Neutralization of TACE α -secretase by ROCKand PDK1 kinases contributes toneurodegeneration in prion diseases: loss-of-PrPC

protective function involved

Mathéa Pietria, Aurélie Alleaume-Butauxa, JulietteEzpeletaa, Vincent Baudouina, Zaira Arellano-Anayaa,Anne Baudrya, Stéphane Haikb, Yannick Baillyc, Jean-Michel Peyrind, Odile Kellermanna, Jean-Marie.Launaye and Benoit Schneidera

aUniversité Paris Descartes, Inserm UMR-S 1124, Paris, France;bINSERM UMR-S 1127, CNRS UMR 7225, Université Pierre-et-MarieCurie, Paris, France; cCNRS UPR 3212, Strasbourg, France; dCNRSUMR 8246, Université Pierre-et-Marie Curie, Paris, France; eINSERMUMR-S 942, Hôpital Lariboisière, Paris, France & Pharma ResearchDepartment, Hoffmann La Roche, Basel, Switzerland

CONTACT Benoit Schneider [email protected]

104 ABSTRACT

ABSTRACT

The α -secretase TACE exerts a protective role at theneuronal cell surface by catalysing the cleavage of sev-eral substrates, including (i) TNF α receptors (TNFRs),which confers physiological sensitivity to TNF α, and(ii) the cellular prion protein PrPC, which limits itsconversion into pathogenic prions PrPSc. We haveassessed whether deregulation of TACE contributes toneurodegeneration in prion diseases.

Using the 1C11 neuronal cell line, primary culturesof neurons and mice infected with prions, we showTACE deficit at the surface of prion-infected cells.Mechanistically, PrPSc promotes the internalization ofTACE into caveolin-1-enriched vesicles, which divertsTACE activity away from TNFRs and PrPC. This ren-ders neurons highly sensitive to TNF α -associatedinflammation and amplifies the production of PrPSc inprion-infected neurons. Whether the neuropathologicaleffect of PrPSc-induced TACE internalization alsodepends on the uncoupling of the α -secretase fromother substrates is currently investigated.

We provide evidence that TACE internalization inprion-infected neurons originates from a loss-of-PrPC

control of TACE localization at the cell surface uponPrPC conversion into PrPSc. The silencing of PrPC inuninfected neurons or the conversion of PrPC intoPrPSc in prion-infected neurons both promote plasmamembrane aggregation of β 1 integrins and a rise of β 1integrin signalling. This leads to the downstream over-activation of the kinase ROCK that complexes withanother kinase, PDK1, and overstimulates PDK1 activ-ity. Overactivated PDK1 then triggers the displacementof TACE from the plasma membrane to caveolin 1-enriched vesicles. The inhibition of ROCK or PDK1 issufficient to target TACE back to the plasma membranewhere TACE recovers its protective activity and therebylimits the production and neurotoxicity of PrPSc or theimpact of a TNF α inflammatory insult. As the phar-macological inhibition of ROCK or PDK1 mitigatesprion diseases in mice, the ROCK-PDK1 duo emergesas a way to combat prion diseases.

192. Prion misfolding activity in ultracentrifugatedsupernatants of ScN2a cell extracts

Margarita Villavedra and David Cullis-Hill

Sylvan Pharmaceutical Pty. Ltd., Sydney, Australia

CONTACT David Cullis-Hill [email protected]

ABSTRACT

Heparan sulphate proteoglycans (HSPG) are involvedin many biological processes. There is evidence thatsuggests they are involved in the conformational con-version of PrPC to the proteinase resistant PrPSC. PrPSC

accumulates, together with HSPG, in the cerebral prionamyloid plaques which are a characteristic feature inthe pathology of the transmissible spongiform encepha-lopathies (TSE). Further, depletion of HSPG in N2acells limited the formation of PrPSC. Also, sulphatedglycans like pentosan polysulfate (PPS) showed capacityto protect PrPC from its conversion to PrPSC in cellculture. Prion proteins have two heparan binding sitesand it has been suggested that the sulphated glycanscompete with the HS that induce the misfolding ofPrPC.

In order to study the role of HSPG in the prionmisfolding process we obtained supernatants of ultra-centrifugated extracts of N2a and ScN2a cells solubi-lized in PBS, 0.5%Triton, 150mM NaCl, 1mM EDTA,pH 7.2. The misfolding capability of these supernatantswere tested by RT-QuIC assay using recombinant 112–231 PrPC (TPrP) or 23-231PrPC (FLPrP).

Our results show that significant misfolding activitypersist in the supernatant after centrifugation at100,000 g for 1h at 4°C. Since PrPSC polymerises intoamyloid rods it is expected to be completely restrictedto the pellet under these centrifugation conditions. TheHSPGs on the other hand are soluble and will remainin the supernatant. Our preliminary results showedthat, particularly using FLPrP, the reactivity of somesupernatants in the RT-QuIC assay can be similar tothat obtained for the original ScN2a homogenates.Although it is not possible to rule out any of thePrPSC being present in the supernatant, its levelswould be expected to be very significantly reduced,which does not correspond to our results obtained inthe quake assay. We propose that HSPG may play arole in the misfolding activity of this ultracentrifugatedsupernatant.

193. Validating the effect of peptide aptamers onthe interaction between Amyloid beta oligomer andPrion protein in vitro and in vivo

Antonia N. Klein, Sabine Gilch

Department of Ecosystem and Public Health, Calgary PrionResearch Unit, Faculty of Veterinary Medicine and Hotchkiss BrainInstitute, University of Calgary, Calgary, Canada

PRION 105

CONTACT Antonia N. Klein [email protected]

ABSTRACT

Introduction: Alzheimer’s disease (AD) is a neurode-generative disease that affects 46.8 Mio people world-wide (estimated in 2015). Aβ monomers aggregates totoxic Aβ oligomers (AβO) and higher aggregates.Studies suggest that the AβO-PrPC-interaction atPrP23-27 (low-affinity side) and PrP95-110 (high-affi-nity side) is required for AβO-induced cell death.Previously, we selected peptide aptamers (PA) bindingto PrPC to inhibit the conformational conversion ofPrPC to infectious PrPSc associated with prion diseases.These PAs consist of a 16 amino acid peptide motifintegrated into bacterial thioredoxin A (trxA) as a scaf-fold protein. One PA, termed PA8, binds to PrPC atposition 100–120. Furthermore, we developed PA8 var-iants, termed 46K, 46Q, and 47H, with single aminoacid replacements to improve the binding affinity. ThePAs increases total PrPC on the cell surface andenhances α-cleavage of PrPC into neuroprotective N1-and C1-fragments. The goal of the study presented herewas to validate these PAs for treatment of AD byinhibiting the AβO-PrPC-interaction.Methods: Synthetic Aβ(1-42) was used to generateAβO, which were characterized by density gradientcentrifugation. AβO-PrPC interaction was analysed byincubating wtN2A cells with FITC-AβO with or with-out PA, PrPC immunostaining and subsequent analysisof FITC-Aβ and PrPC colocalization. Toxicity inducedby the AβO-PrPC-interaction was analysed by MTTassays and Western blot analysis of active Caspase 3.In the ongoing in vivo study, 7 weeks old 5xFAD miceare treated with PA for 12 weeks intraventricularlyusing osmotic Alzet® pumps. Behavioural tests likenovel object recognition test and Morris water mazewill be performed within the last treatment week.Results: The here used PAs inhibited AβO-PrPC-inter-action. Treatment of wtN2A cells with AβO led to areduced cell viability of 34 ± 7%. Concomitant treat-ment of wtN2A cells with PA8 and 46K for 24 hresulted in a significant increase of cell viability to44 ± 8% and 48 ± 16%, respectively. Treating SH-SY5Y cells (not detectable PrPC levels) with PAs andAβO, followed by MTT assay confirmed that theenhanced cell viability is PrPC dependent. Westernblot analysis of active Caspase 3 proved reducedCaspase 3 activation after treatment of wtN2A cellswith PAs. Whether these effects ultimately result inmemory improvement will be evaluated through thein vivo studies.

Conclusion: In conclusion, the here used PA were ableto inhibit the AβO-induced toxicity in vitro. Thesefindings indicate that PAs are interesting candidatesfor the treatment of both prion diseases and AD.

194. The photodynamic inactivation reduces prioninfectivity in phthalocyanine treated RML brainhomogenate

Marie Kostelanskaa, Zdenka Backovska Hanusovaa,Jakub Soukupa, Radoslav Matejb and Karel Holadaa

aInstitute of Immunology and Microbiology, First Faculty ofMedicine, Charles University and General University Hospital inPrague, Czech Republic; bDepartment of Pathology and MolecularMedicine, Thomayer Teaching Hospital, Prague, Czech Republic

CONTACT Jakub Soukup [email protected]

Introduction: Prions are highly resistant to conven-tional sterilization procedures and currently recom-mended inactivation methods include treatment withhighly corrosive chemicals like 2% NaClO or 1MNaOH. Such harsh treatment is incompatible with anumber of delicate medical tools. We have previouslydemonstrated that nontoxic disulfonated hydroxyalu-minum phthalocyanine (AlPcOH(SO3)2) can be usedto initiate photodynamic inactivation (PDI) of prionsat ambient pressure and temperature [1]. The aim ofthis study was to demonstrate the effectiveness of PDIof prions by bioassay in laboratory mouse.Methods: The PDI was performed on 1% RML or unin-fectious CD1 brain homogenates (BH) treated with 25 µgml−1 of AlPcOH(SO3)2 and exposed to red light (18 min,4 × 2.5W LED). Control RML inoculum was preparedsimilarly but was kept in the dark. In mouse bioassay,CD1 female mice (n = 10) were intracerebrally inoculatedwith a 25-µl aliquot of AlPcOH(SO3)2-light treated RMLBH or control RML BH containing AlPcOH(SO3)2 butnot irradiated by light. Other controls included mice(n = 5) inoculated with untreated, uninfectious CD1 BHor CD1 homogenate containing AlPcOH(SO3)2, eitherirradiated or nonirradiated. For comparison groups ofmice (n = 5) were inoculated with 10-fold dilutions ofRML BH (10−2 to 10−6). The animals were sacrificed afterreaching the terminal disease phase. The presence ofPrPres in mouse brains was detected by WB and evalua-tion of PrPres distribution in brains by immunohisto-chemistry is ongoing.Results: Mice inoculated with PDI-treated RML BH (at a10−2 dilution) survived significantly longer (204 ± 23 days)than mice inoculated with the homogenate containing an

106 ABSTRACT

identical amount of AlPcOH(SO3)2, that was not exposedto light (165 ± 11 days). Their survival was also significantlylonger than that of mice inoculated with straight 10−2, 10−3,and 10−4 dilutions of the RML BH, who survived 169 ± 11,165 ± 12, and 176 ±15 days, respectively. The transmissionrate was 100% in all groups ofmice including serial dilution(10−2 to 10−6). A comparison of survival time based on theregression line suggested a decrease in the prion infectivitytiter of the RMLBHofmore than four orders of magnitudeafter PDI treatment.Conclusions: Our results suggest that PDI withAlPcOH(SO3)2 leads to substantial decrease (~4 log10)of prion infectivity in RML BH. The PDI of prions bylight induced photocatalytic activity of phthalocyaninesrepresents promising way of prion decontamination.

KEYWORDS: Prion; phthalocyanine; photodynamicinactivation; decontamination; mouse bioassayThe project was supported by AZV NV18-04–00179.

Reference

[1] Janouskova et al., J Gen Virol. 2012; 93(Pt11): 2512–2517.

195. When anchorage determines fate: How theGPI-anchor signal sequence impacts on prionprotein biology and prion disease in mice

Berta Puiga,b, Hermann C. Altmeppena, LuiseLinsenmeiera, Karima Chakrouna, Ulrike K. Piontekc,Jörg Tatzeltc,d, Clive Batee, Tim Magnusb and MarkusGlatzela

aInstitute of Neuropathology; bDepartment of Neurology,Experimental Research in Stroke and Inflammation (ERSI),University Medical Center Hamburg-Eppendorf (UKE), Hamburg,Germany; cAdolf Butenandt Institute, Ludwig MaximiliansUniversity, Munich, Germany; dInstitute of Biochemistry andPathobiochemistry, Biochemistry of Neurodegenerative Diseases,Ruhr University, Bochum, Germany; eDepartment of Pathologyand Pathogen Biology, Royal Veterinary College, Hawkshead Lane,North Mymms, Herts, UK

CONTACT Hermann C. Altmeppen [email protected]

ABSTRACT

Correct anchoring and localization of the prion pro-tein (PrP) at the neuronal surface are essential for itsprocessing, functions and roles in disease. Severalstudies have revealed a compensatory network regu-lating PrP membrane homeostasis, including proteo-lytic or extracellular vesicle release, andinternalization followed by retrograde transport anddegradation. In this study however, we asked if theGPI-anchor signal sequence (GPI-SS) -though not

part of the mature protein- affects composition, traf-ficking, membrane localization and, hence, biology ofPrP in the first place, and whether alterations in thatregard would impact on prion disease in particular.We generated different lines of transgenic miceexpressing a mutant PrP (PrPCGPIThy-1) containingthe GPI-SS of the neuronal protein Thy-1, a residentof dense cores of lipid rafts. This modification indeedresulted in a different GPI-anchor composition (nowlacking the rare mammalian sialic acid typical forPrP), altered sorting in polarized cells (such as pri-mary neurons), and reduced proteolytic shedding bythe metalloprotease ADAM10. When inoculated witheither RML or 22L prions, survival of mice was sig-nificantly prolonged for all transgenic lines, despitesimilar or even higher expression levels of generallysignalling- and misfolding-competent PrPCGPIThy-1compared to wild-type controls. Fittingly, MAPkinase signalling as well as glial activation and spon-giform lesions were reduced in brains of our trans-genic mice. Using PrP as a model, this is the first invivo study demonstrating that the GPI-SS determinesthe later remodelling and composition of a givenprotein`s GPI-anchor and sorting, with likely conse-quences for its biology. Besides drastic effects ondifferent pathomechanistic aspects underlying thecourse of prion diseases, our findings may also havewider implications for the field of GPI-anchorbiology.

KEYWORDS: ADAM10; glycosylphosphatidylinositol(GPI)-anchor; lipid rafts; prion protein; shedding; sig-nalling; sorting

Reference

[1] Puig, Altmeppen et al., PLoS Path. 2019.

196. Reverting prion disease in neuronalprogenitor cells as a cell replacement therapy

Arielle Hay, Lindsay Parrie, Glenn C. Telling, MarkZabel and Julie A. Moreno

Prion Research Center, Department of Microbiology, Immunology &Pathology, Colorado State University, Fort Collins, CO, USA

CONTACT Arielle Hay [email protected]

ABSTRACT

We propose the use of gene edited stem cells astherapy for neurodegenerative diseases. Olfactoryneuronal progenitors (ONPs) can differentiate intoneurons in adulthood. We hypothesize they will

PRION 107

regenerate neurons that have been lost due to aggre-gation of disease- associated proteins. Mesenchymalstem cells (MSCs) can be derived from adipocytes ofadult mice and further differentiated to neural stemcells (NSCs), which we plan to use as an alternativeto ONPs. These therapies will be modelled by prioninfected mice, which display the typical features ofneurodegenerative disease, including clinical signsand loss of neurons. Prion diseases are the result ofmisfolding of the prion protein (PrP), which is highlyexpressed in neurons. PrP knockout mice are resis-tant to prion diseases. Two populations of ONPs willbe made using CRISPR/Cas9 gene editing to deletethe prnp gene in N2a cells and WT ONPs. These cellswill be subjected to prion cell assays to determine ifthey can be infected. The second population of ONPswill be from mice with a mutation associated withthe human prion disease Gerstmann–Sträussler–Scheinker syndrome (GSS), which is modelled inmice by a proline to leucine switch at codon 101 ofmoPrP. We plan to use CRISPR/Cas9 to revert thismutation. Alternatively, we plan to edit the prnp genein M/NSCs so they express a secretable, dominant-negative PrP. Single cell sorting will be used to pro-duce homogenous populations of edited cells, whichwill be sequenced to identify relevant mutations. Wewill engraft gene edited ONPs using stereotacticinjection and M/NSCs using nasal instillation intothe brains of prion infected WT and GSS transgenicmice [1]. We hypothesize that these cells will resistprion infection. ONPs will migrate throughout thebrain to restore damaged neurons and M/NSCs willpopulate the olfactory bulb and secrete dominantnegative PrP and anti-inflammatory cytokinesthroughout the brain.

197. Proteolytic shedding of PrP can bemanipulated in a substrate-specific manner bycertain PrP-binding ligands

Luise Linsenmeiera, Behnam Mohammadia, MohsinShafiqa, Simrika Thapab, Karl Frontzekc, Berta Puiga,Chaoyang Lid, Michaela Schweizere, Tania Massignanf,Emiliano Biasinif, Antonia Kleinb, Sabine Gilchb, LucaVaranig, Hermann Schatzlb, Adriano Aguzzic,Hermann C. Altmeppena* and Markus Glatzela*aInstitute of Neuropathology, University Medical Center HH-Eppendorf, Hamburg, Germany; bCalgary Prion Research Unit,Faculty of Veterinary Medicine, University of Calgary, Canada;cInstitute of Neuropathology, University Hospital Zurich,Switzerland; dWuhan Institute of Virology, Chinese Academy ofScience, Beijing, China; eCenter for Molecular NeurobiologyHamburg, Morphology and Electron Microscopy Unit, University

Medical Center HH-Eppendorf, Hamburg, Germany; fDepartmentof Cellular, Computational and Integrative Biology (CIBIO),University of Trento, Italy; gInstitute for Research in Biomedicine,Università della Svizzera italiana, Bellinzona, Switzerland

*These authors contributed equally to this work.

CONTACT Luise Linsenmeier [email protected]

ABSTRACT

The prion protein (PrPC) impacts on development andmaintenance of the nervous system and plays a deleter-ious role in fatal and progressive prion diseases.Moreover, in other neurodegenerative proteinopathiesit acts as a receptor for toxic oligomers such as Aβ inAlzheimer’s disease (AD). PrPC is released from the cellsurface into the extracellular space by the metallopro-tease ADAM10. We have shown that this cleavage sig-nificantly impacts on prion disease, and soluble PrPfragments may be key in understanding the multitudeof functions suggested for PrPC, such as in myelinmaintenance, neuroprotection, synapse formation orneuroimmune crosstalk.

We expect the ADAM10-mediated shedding of PrPC

to have therapeutic potential against neurodegenerativeproteinopathies. However, a substrate-specificapproach to stimulate this cleavage would likely besuperior to targeting of the protease, given the plethoraof ADAM10 substrates throughout the body and, thus,the risk of severe side-effects. We found that severalPrP-directed antibodies significantly increase sheddingin different cell culture models and organotypic slicecultures. Crosslinking of PrP is not a prerequisite sincetreatment with single chain antibodies and other PrP-binders likewise resulted in increased cleavage. We thusassume that binding of certain ligands to PrP per sestimulates its shedding. While we also identified someantibodies that blocked shedding, one particular anti-body (likely due to its unique binding characteristics)led to strong surface clustering, fast internalization anddegradation of PrP rather than to proteolytic releasefrom the membrane.

Of note, PrP-directed antibodies or compounds havebeen experimentally suggested or are currently tested asa treatment option in fatal neurodegenerative proteino-pathies. It remains to be investigated whether stimu-lated shedding is (at least partially) underlying theprotective mechanism. In contrast, given the recentlysuggested harmful roles of shed PrP in brain cancer andHIV-associated neuropathogenesis, the shedding-inhi-biting ligands identified in our study may also holdtherapeutic potential in pathological conditions affect-ing the brain.

108 ABSTRACT

198. Predicting Blood-Brain Partitioning and SkinPermeability using 3D-RISM-KH Theory BasedSolvation Energy Descriptors

Andriy Kovalenko

Department of Mechanical Engineering, University of Alberta, 10–203 Donadeo Innovation Centre for Engineering, Edmonton,Alberta, Canada; Nanotechnology Research Centre, Edmonton,Alberta, Canada

ABSTRACT

Compartmentalization of drug molecules betweenplasma and brain is important for desired activity.The difficulty in obtaining the blood-brain permeabilityof drug (like) substances experimentally resulted in thedevelopment of several theoretical quantitative struc-ture activity relationships (QSAR) towards predictingcapability of a given substrate to pass across tight junc-tion, known as the blood-brain barrier, both qualita-tively and quantitatively. We have developed a novelmethod based on the three-dimensional reference inter-action site model with the Kovalenko-Hirata closurerelation (3D-RISM-KH) molecular solvation theoryfor predicting blood-brain barrier permeability coeffi-cients of molecules in diverse structures with a mini-mum set of descriptors derived from solvationenergetics [1]. Our findings reveal the importance ofsolvation free energy based descriptors in quantitativelymodelling blood-brain barrier permeability. Further, weuse the state-of-the-art molecular solvation theory topredict skin permeability of a large set of compoundswith experimental logKP [2]. We obtained encouragingresults pointing to applicability of the model usingstatistical physics based 3D-RISM-KH theory for solva-tion free energy calculations as a primary descriptorwith relative mean square error of 0.77 units.

References

[1] Roy D, Hinge VK, Kovalenko A. Predicting blood-brain partitioning of small molecules using a novelminimalistic descriptor based approach via the 3D-RISM-KH molecular solvation theory. ACS Omega.2019 (accepted).

[2] Hinge VK, Roy D, Kovalenko A. Predicting skin per-meability using the 3D-RISM-KH theory based solva-tion energy descriptors for a diverse class ofcompounds. J Pharm Sci, 2019 (submitted).

199. Chronic wasting disease transmission studiesto non-human primates and transgenic mice

Brent Race, Katie Williams, Christina D. Orrù, AndrewG. Hughson, Lori Lubke and Bruce Chesebro

National Institute of Allergy and Infectious Diseases, Laboratory ofPersistent Viral Diseases, Rocky Mountain Laboratories, Hamilton,MT, USA

CONTACT Brent Race [email protected]

ABSTRACT

Introduction: Chronic wasting disease (CWD) is theonly prion disease affecting free-ranging animals,reported in North America, South Korea and Norway.Unlike in most other prion disease CWD agents areshed in blood, urine and feces which most likely con-tribute to the horizontal transmission between cervidspecies. The developments of amplification-based seed-ing assays have been instrumental in the detection oflow levels of prions in clinical samples. Using real-timequaking-induced conversion (RT-QUIC), we estab-lished an ultrasensitive detection method forPrPCWD in the urine from CWD-infected sequentiallysampled transgenic mice overexpressing elk prion pro-tein (TgElk mice). In addtion, RT-QUIC was per-formed in the kidney and brain of theses mice modelto trace abnormal prion.Materials and Methods: 44 brain and kidney, urine sam-ples from sequentially collected from CWD-infectedTgElk mice (TgElk CWD) were stored at -80℃. In brainand kidney, 10% (w/v) homogenate was prepared in 0.9%sterilized saline. In urine 100uL of each sample was mixedwith 10uL 2.8% sodium phosphotungustic acid (NaPTA)and incubated for 1hr at 37℃ with shaking at 1,350 rpm.Samples were centrifuged for 30min at 16,100g. The pelletwas resuspended in 10uL of 0.1% SDS/PBS for 30min at55℃. RT-QUIC reactions were set up in 96-well clearbottom optic plates and consisted of 98uL RT-QUICbuffer [final concentrations of 1XPBS, 1mM EDTA,10uM Thioflavin, 300mM NaCl buffer and 0.1mg/mlrecombinant Syrian hamster recombinant protein (23-231) and 2uL of sample. The RT-QUIC assay was per-formed on a FLUOstar Omega fluorescence plate readerthat was preheated to 42℃ for 60hr with 90sec shaking at700rpm followed by 1min incubation.Results: Five randomly selected mice were sequentiallyculled on every 15 days from 30dpi to 120dpi duringCWD infected TgElk mice reached terminal stage.Rough hair coats among clinical signs were showedfrom 90 dpi. PrPCWD in the brain in TgElk CWD wasdetectable persistently from early stages (30dpi), and inthe kidney PrPCWD was also detectable in clinical andterminal stages (90 dpi and 120dpi). PrPCWD in theurine in TgElk CWD reached the highest levels at120dpi. NaPTA/RT-QUIC was applied to measurePrPCWD in urine samples collected on every 15 daysfrom 30dpi to 120dpi when CWD infected TgElk mice

PRION 109

reached terminal stage. PrPCWD in the urine in TgElkCWD reached the highest levels at 90dpi. PrPCWD wasalso detectable in late and terminal stages (120dpi).Conclusions: We demonstrate that CWD prions can bedetected by RT-QUIC or NaPTA/RT-QUIC in the brain,kidney and urine of TgElk mice at the early and terminalstages of disease. Based on these data, we suggest thatPrPCWD is excreted into only urine until 90 dpi and thenslowly accumulated in kidney. Our results can be used indesigning future study of CWD pathogenesis in TgElkmice.KEYWORDS: Cynomolgus macaques; squirrel mon-keys; non-human primate; transgenic mice; CWD;RT-QuIC; prion; cross-species transmission; barrier;chronic wasting disease

References

[1] Henderson DM et al Rapid antemortem detection ofCWD prions in deer saliva PLOS one 2013, 397, 626-630.

[2] Nagooka K, Yoshika M, Shimozaki N. et al. Sensitivedetection of scrapie prion protein in soil. BiochemBiophys Res Commun. 2010 e74377 1-12.

200. DNA methylation array based machinelearning classification of human prion disease

Fernando Guntoro, Luke Dabin, John Collinge,Emmanuelle Vire, and Simon Mead

MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, UK

CONTACT Fernando Guntoro [email protected]

ABSTRACT

Background: Variant Creutzfeldt-Jakob disease (vCJD)was caused by the widespread dietary exposure of theUnited Kingdom (UK) and other populations to bovinespongiform encephalopathy (BSE) prions. Unlike otherforms of CJD, abnormal prion proteins (PrP) in vCJDare readily detectable by conventional histopathologyoutside the central nervous system, particularly in lym-phoreticular system (LRS) tissues such as tonsillar andappendix tissues. To date, six prevalence studies havebeen conducted; the fifth (Appendix II) and sixth(Appendix III) studies reported abnormal PrP immu-noreactivity in 16 out of 32,441 appendix samples and 2out of 14,692 samples, respectively, indicating a surpris-ingly high prevalence abnormal PrP. Given the 178vCJD cases hitherto reported in the UK, there appearsto be a discordance between the population prevalencedata and the number of observed clinical cases, whichchallenges the assumption of a perfect association

between PrP deposition in LRS tissues and vCJD dis-ease. To provide insight into this discordance, we aimto develop a classifier based on epigenomic profiles thatdistinguishes prion infection from healthy controls.Materials and Methods: A unique archive of fixed andfrozen LRS tissues including tonsil and appendix fromvCJD patients, blood and brain samples from a range ofhuman prion disease patients. Samples have been age/sex-matched to controls not diagnosed with neurode-generative disease. For DNA methylation profiling,extracted genomic DNA has been subjected to bisulfiteconversion and, subsequently, hybridized to theIllumina 450K or 850K EPIC arrays. Training of adeep learning neural network model was performedusing these samples.Results: Initial results explored the diagnosticpotential of an Illumina 450K dataset using deeplearning neural network classifier. This DNA methy-lation study was conducted using whole bloods from106 healthy controls and 114 sCJD patients (age/sex-matched). Using data from the 1000 most sig-nificantly altered methylation loci, this data wasrandomized and partitioned into training and testset of 1:1 case-control ratio. After 10-fold validation,the model had an accuracy of 85.64% (± 1.14%)with an upper limit of 90.00% accuracy. The trainedneural network model demonstrated a better perfor-mance compared to a basic Random Forest classifierwith an AUC of 0.965 and 0.841, respectively.Conclusions: As a proof of concept, we have developedan accurate classification tool using relatively smallnumbers of differentially methylated loci in blood. Weare extending these results to a wider panel of neuro-degenerative dementias and LRS tissues.

KEYWORDS: Epigenetics; EWAS; DNA methylation;Variant Creutzfeldt-Jakob; Machine learning;Appendix; Tonsil; Lymphoreticular tissue

201. TDP-43 induces SOD1 aggregation andtoxicity via tryptophan interactions that present anovel drug target

Michèle G. DuVala, Edward Pokrishevskyb, Vijaya K.Hingec,d, Jenna Bratvolda, Richard Kanyoa,e, NatalieSnydera, Nikolay Blinovc,d, Andriy Kovalenkoc,d, NeilR. Cashmanb and W. Ted Allisona,e,f

aDepartment of Biological Sciences, University of Alberta, Edmonton,AB, Canada; bDjavad Mowafaghian Centre for Brain Health,University of British Columbia, Vancouver, BC, Canada; cNationalInstitute for Nanotechnology, Edmonton, AB, Canada; dDepartmentof Mechanical Engineering, University of Alberta, Edmonton, AB,Canada; eCentre for Prions and Protein Folding Diseases, University

110 ABSTRACT

of Alberta, Edmonton, AB, Canada; fDepartment of Medical Genetics,University of Alberta, Edmonton, AB, Canada

CONTACT Michèle G. DuVal [email protected]

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastatingmotor neurodegenerative disease which features adiversity of protein inclusions. Aggregates of superox-ide dismutase 1 (SOD1) or of TAR-DNA binding pro-tein-43 (TDP-43) have been identified in familial andsporadic cases. Misfolded wildtype SOD1 can spread ina prion-like fashion, evidenced by induction of wildtypeSOD1 misfolding by mutant SOD1 or by TDP-43 var-iants, as observed in cells and cultured neurons [1–3].The sole tryptophan in SOD1 at position 32 (W32) isinstrumental in SOD1 misfolding [1,2,4], and wehypothesize that TDP-43 also induces SOD1 misfoldingvia interactions between their respective tryptophanresidues. We measured the contribution of these tryp-tophans to SOD1 aggregation in vitro and toxicity invivo by expressing variants with combinations of tryp-tophan substitutions.

In HEK293 cells, TDP-43 lacking all six tryptophansinduced little SOD1 inclusion formation, which wasreplicated with TDP-43 variants lacking two specifictryptophans. In a zebrafish model of motor neurondisease [5–7], disturbed motor axon morphologycaused by TDP-43 and SOD1 co-expression was simi-larly rescued, with congruent results for substitutions ofspecific tryptophans in TDP-43. Thus TDP-43 caninduce SOD1 aggregation and toxicity via interactionsbetween tryptophans on the two proteins, which mayoccur under pathological conditions and therebyadvance disease. Similarly, mutating W32 in thehuman SOD1 delivered to zebrafish lead to significantreduction of motor neuron phenotypes (vs wildtypeSOD1), including preservation of axon morphologyand of motor output (swimming).

Candidate therapeutic compound 5’-fluorouridine,known to bind at the W32 region of SOD1 [8],successfully rescued cross-seeding of SOD1 by TDP-43 and subsequent motor neuron toxicity. Next, toidentify additional compounds that would bind atW32, high throughput virtual and pharmacophorescreening was performed with modelling of theW32 binding site and screening of FDA-approveddrugs that closely match key features of 5’-fluorour-idine. From among top-ranked compounds, telbivu-dine significantly rescued SOD1-induced motorneuron phenotypes [9]. In conclusion, we foundthat tryptophan 32 in SOD1 is a significant contri-butor to SOD1 aggregation and toxicity, including via

novel TDP-43-SOD1 interactions. These interactionsrepresent new pharmacologic targets and we identi-fied FDA-approved compounds, predicted to interactwith SOD1 at W32, that are effective in preventinginclusion formation and toxicity.

KEYWORDS: SOD1; TDP-43; prion-like; ALS; trypto-phan; 5’-fluorouridine; telbivudine; zebrafish

References

[1] Grad et al., PNAS. 2011;111:3620–3625.[2] Taylor et al., J Biol Chem. 2007;282:16329–16,335.[3] Pokrishevsky et al., Sci Rep. 2016;6:22155.[4] Pokrishevsky et al., Sci Rep. 2018; 8:15590.[5] Sakowski et al., Mol Neurodegener. 2012;7:44.[6] DuVal et al., PLoS One. 2014;9:e89183[7] Edens et al., eLife. 2017; 6:e25453[8] Wright et al., Nat Commun. 2013;4:1758[9] DuVal et al., Neurobiol Dis 2019; 124: 297–310

202. Competitive ELISAs to determine PrPSc

content in crude homogenates without the need fordenaturation or protease treatments

Xinli Tanga,b, Andrew Fanga,b, Leonardo M. Corteza,c,Camilo Duque Velásqueza,d, Claudia Y. Acevedo-Morantesa,b, Brian Tancownya,b, Valerie L. Sima,c,Judd Aikena,e, Debbie McKenziea,d and Holger Willea,b

aCentre for Prions and Protein Folding Diseases, University ofAlberta; bDepartment of Biochemistry; 3Department of Medicine;dDepartment of Biological Sciences, eDepartment of Agricultural,Food, and Nutritional Science, Edmonton, Alberta, Canada

CONTACT Xinli Tang [email protected]

ABSTRACT

Background: The lack of readily accessible, PrPSc-spe-cific monoclonal antibodies has hampered the detectionof PrPSc in brain homogenates and other samples. Thepresence of PrPC and other, non-infectious PrP con-formers confounds diagnostic assays that lack specifi-city for native PrPSc. Currently used diagnostics oftenrely on the partial protease resistance of PrPSc, which isused to differentiate PrPSc from PrPC and other PrPconformers. Furthermore, the aggregated nature ofPrPSc obscures many epitopes, making denaturation ofPrPSc a prerequisite for many detection assays.Together these requirements affect the specificity andsensitivity of the detection assays and increase thenecessary labour, time, and resources to determine ifa sample contains PrPSc.Materials andMethods:We immunizedmicewith a struc-tural and immunological mimic for PrPSc, which was basedon the fungal HET-s prion domain as a scaffold. At the end

PRION 111

of the immunization schedule we used their spleens togenerate hybridomas for the production of monoclonalantibodies. Here, we tested the resulting antibodies in con-ventional and competitive ELISAs for their ability to detectnative PrPSc from different prion isolates and to distinguishPrPSc from PrPC and other PrP conformers.Results: Immunization with our newly developed, struc-tural mimic gave rise to a PrPSc-specific immune responsein 8/8 mice. The resulting monoclonal antibodies (IgGsand IgMs) recognized native PrPSc in crude brain homo-genates without any pre-treatment, but did not bind toPrPC, recombinant PrP, denatured PrP, unseeded PrPamyloid, or any linear peptide of PrP. We tested a varietyof assay formats and obtained the most reliable resultsusing a competitive/inhibition ELISA. Here, our originalantigen was used to coat the assay plates and to competewith PrPSc in the samples (typically crude brain homo-genate) for binding of unconjugated antibodies. A con-ventional detection system using a secondary antibody(HRP-goat-anti-mouse IgG or IgM) generated the finalcolorimetric readout. The assay recognized native PrPSc

from all prion isolates tested, including CWD, BSE (C-,H-, and L-type), scrapie (RML), TME (Hyper andDrowsy), sCJD, vCJD, fCJD, FFI, and GSS, indicating itsability to detect a widely-shared PrPSc-specific epitope.Conclusions: Our new, PrPSc-specific monoclonal anti-bodies allowed the development of a competitive ELISAfor the detection of native PrPSc without protease ordenaturant pretreatment. Furthermore, the broad spe-cificity of these antibodies opens the possibility todevelop a widely applicable and sensitive diagnostictool for the direct detection of PrPSc.

203. Seeding of a fast acting Aß peptideaccelerates AD-like pathology in APPNL-G-F mousemodel with no impairment in learning and memory

Sean G. Lacoursiere, Majid H. Mohajerani and RobertJ. Sutherland

Canadian Centre for Behavioural Neuroscience, University ofLethbridge

CONTACT Sean G. Lacoursiere [email protected]

ABSTRACT

Alzheimer’s disease (AD) is a neurodegenerativedementia characterized by the stereotypical conforma-tional misfolding of proteins. Of specific interest isamyloid-ß (Aß) due to its prion-like properties evi-denced by the seeding of Aß accelerating plaquedeposition in brain. However, the role of Aß in ADremains unclear. Many hypothesize that increasing Aßimpairs cognition; and therefore, accelerating Aß

pathology should increase the extent of cognitivedecline to the point of dementia. We performed bilat-eral intracerebral microinjections of a fast-acting Aßpeptide into the medial entorhinal cortex of theknock-in APPNL-G-F mouse model of AD at 2 monthsof age. We found deposition of Aß plaque occurredsooner and to a greater extent compared to controls butwithout obvious impairment in cognition or overallhealth. We found that at 6 months the APPNL-G-F

mice showed similar memory performance in a no-platform Morris Water Task probe test compared tocontrols. Immunohistochemical labelling showed dra-matically increased 82E1 and Iba1 label in these mice1 month following injection, indicating increased Aßand microgliosis pathology, respectively. Our resultsreplicate previous reports of the seeding properties ofAß and its prion-like behaviour. They also supportnotions that Aß may not be sufficient to cause cognitivedecline in Alzheimer’s disease. The Aß pathology inAD may be a response to some other trigger. Theseeding properties of Aß may be an adaptive responseto control some other process.

204. Alteration to the prion protein’s centralregion sequence retains structure but results intoxicity

Graham P. Rosemana, Bei Wub, Mark Wadolkowskia,David A. Harrisb and Glenn L. Millhausera

aUniversity of California Santa Cruz; bBoston University School ofMedicine

CONTACT Graham P. Roseman [email protected]

ABSTRACT

The central region (CR) of the prion protein (PrPC) isone of the most highly conserved sequences acrossspecies. Deletion of the entire prion protein gene inmice is fairly tolerated, but deletion of just the CR(ΔCR: 105–125) causes neonatal fatality in mice [1]and toxicity in cell culture as seen by spontaneouscurrents using patch clamp electrophysiology experi-ments. There is a great interest in understanding thetoxicity of this deletion mutant because the neurotoxi-city has similarities to that of classic prion diseasesexcept without the aggregation of PrPC. Biophysicalstudies have shown that ΔCR causes two main changes:weakening of the metal-driven cis interaction betweenthe N- and C- terminal domains [2] and deletion of theregion of the protein where regulatory proteolysisoccurs, termed α-cleavage. What is not known iswhether the neurotoxicity that ΔCR causes is a resultof a weakened cis interaction or elimination of

112 ABSTRACT

proteolytic sites. This study aims to ask that question byusing a designed PrPC construct that replaces the cen-tral region with a flexible glycine-serine (GS) rich linkerthat retains the metal-driven cis interaction of wild typePrPC while blocking the cleavability of this region.Results from the GS linker show that the cis interactionis retained as tested by NMR, α-cleavage was blocked invitro and in cell culture, and toxicity was seen usingelectrophysiological experiments. To test if the toxicitygenerated from the GS linker construct was due fromeliminating α-cleavage, a rescue construct was designedby reintroducing the canonical α-cleavage sequenceinto the GS linker backbone. α-cleavage was success-fully reintroduced and the metal-driven cis interactionwas retained. However, when this construct was testedusing the electrophysiology experiments, spontaneouscurrents still persisted, indicating that there was stilltoxicity. Overall these results implicate that thesequence of the central region is extremely importantfor the structure and function of the prion protein, andwhen deleted or mutated, spontaneous neurotoxicityoccurs.

KEYWORDS: Metal binding; copper; cleavage; NMR;electrophysiology

References

[1] Li et al., EMBO. 2007;26:548–558.[2] Wu et al., eLife. 2017;6:e23,473

205. Deep learning-based pre-mortem diagnosticmethods for sporadic Creutzfeldt-Jakob diseaseusing multiple CSF biomarkers

Sol Moe Leea,b, Jae Wook Hyeona, Soo-Jin Kimb,Heebal Kimb, Ran Noha, Seonghan Kima, YeongSeon Leea, Sung Soon Kima and Su Yeon Kima

aDivision of Bacterial Disease Research, Center for InfectiousDiseases Research, Korea National Institute of Health, Centers forDisease Control and Prevention, Cheongju-si, Chungcheongbuk-do,South Korea; bDepartment of Agricultural Biotechnology andResearch Institute of Agriculture and Life Sciences, Seoul NationalUniversity, Seoul, South Korea

CONTACT Sol Moe Lee [email protected]; Su Yeon Kim [email protected]

ABSTRACT

Creutzfeldt-Jakob disease (CJD) is a fatal neurodegen-erative disorder which is characterized by the abnormalaccumulation of misfolded prion protein (PrPSc)

affecting the central nervous system. The diagnosis ofCJD can only be confirmed by abnormal protease-resistant prion protein accumulation in post-mortembrain tissue. Sporadic CJD (sCJD) is the most commonsubtype, and the relationships between sCJD and cere-brospinal fluid (CSF) proteins such as 14–3-3, tau, andα-synuclein (a-syn) have been investigated for theirpotential values in pre-mortem diagnosis. Deep learn-ing (DL), a machine learning (ML) based on neuralnetworks, has provided a powerful remedy for analys-ing large-scale data in diverse fields such as computervision, natural language processing, and signal model-ling for several years. Recently, it is reported that DLhas attracted large attention in neurodegenerative dis-ease research as well. Here, ML and DL analysis wereperformed using multiple CSF biomarkers related withneuronal diseases including CJD and Alzheimer’s dis-eases. We performed ML analysis including decisiontrees and SVMs using WEKA (Waikato Environmentfor Knowledge Analysis 3.8.2), and DL analysis wasconducted with Keras neural network library.Enzyme-linked immunosorbent assays were performedon phospho-tau (p-tau), total-tau (t-tau), a-syn, and β-amyloid (1-42), and western blot analysis was per-formed for 14–3-3 protein from CSF samples of 49sCJD and 256 non-CJD Korean patients, respectively.The best performing ML model demonstrated 78.85%accuracy. The deep neural network structure whichshowed the best DL performance was comprised ofone input, five hidden, and one output layers, with20, 40, 30, 20, and 12 hidden unit numbers per hiddenlayer, respectively. For the analysed CSF biomarkers,the DL model demonstrated 90.38% accuracy, 83.33%sensitivity, and 92.5% specificity for the three-proteincombination of t-tau, p-tau, and a-syn. DL-aided pre-mortem diagnosis may provide a suitable tool for dis-criminating CJD patients from non-CJD patients. Forfurther study, we plan to perform DL analysis usingstacked sample sizes via ELISA and western blot analy-sis to establish a DL model with greater high perfor-mance and classification accuracy.

206. Characterizing the molecular environment ofthe bank vole prion protein using massspectrometry

Hamza Arshada,b, Declan Williamsa, LechKaczmarczykc, Walker Jacksonc, Gerold Schmitt-Ulmsa,d, Joel Wattsa,b

aTanz Centre for Research in Neurodegenerative Diseases, Toronto,Canada; bDepartment of Biochemistry, University of Toronto,Toronto, Canada; cWallenberg Center for Molecular Medicine,

PRION 113

Linköping University, Linköping, Sweden; dDepartment ofLaboratory Medicine and Pathobiology, University of Toronto,Toronto, Canada

CONTACT Hamza Arshad [email protected]

ABSTRACT

Background: The bank vole prion protein (BVPrP) isable to replicate a wide range of prion strains fromdifferent species and has been characterized as a uni-versal acceptor of prions [1,2]. The sequence of matureBVPrP differs from that of mouse PrP (MoPrP) at onlyeight different amino acid positions. We hypothesizedthat these unique residues may allow BVPrP to tailor itsmolecular environment through novel protein-proteininteractions. To address this issue, we generated knock-in (ki) mice that express BVPrP (M109 isoform) atphysiological levels in the brain. The PrP interactomesin ki mice expressing BVPrP and wild-type miceexpressing MoPrP were compared using quantitativemass spectrometry.Materials and Methods: BVPrP ki mice, wild-typeC57Bl/6 mice, and PrP knockout mice were subjectedto time-controlled transcardiac perfusion crosslinking(tcTPC) and then PrP- containing protein complexeswere isolated from the brain by immunoprecipitation[3]. Triplicate samples were analysed by quantitativemass spectrometry to permit a direct comparison ofthe interactomes of MoPrP and BVPrP. The interac-tome analysis was conducted twice, using two differentanti-PrP antibodies recognizing distinct epitopes, toensure that all relevant interactions were captured.Results: The molecular environment of BVPrP wasvery similar to that of MoPrP. Many known MoPrPinteractors were found in our experiments, includingNCAM, 4F2, basigin, as well as subtle interactors suchas ZIP6 and ZIP10. We found that there was significantoverlap between the top interactors found in the BVPrPand MoPrP samples. However, we also identified sev-eral proteins that were reproducibly enriched in thesamples derived from BVPrP ki mice. These hits arecurrently being validated using cultured cell models.Conclusions: We conclude that the molecular environ-ments of BVPrP and MoPrP in the brains of mice arevery similar. Nonetheless, several proteins appear topreferentially interact with BVPrP. These proteinsmay help to explain the unique behaviour of BVPrP.

KEYWORDS: Bank vole; mass spectrometry; interac-tome; species barrier

References

[1] Watts et al., PLoS Pathog. 2014;10(4):e1003990.

[2] Agrimi et al., PLoS Pathog. 2008;4(7):e1000113.[3] Schmitt-Ulms et al., Nat Biotech. 2004;22(6):724–731.

207. Neurodegeneration in the enteric nervoussystem in an MPTP induced model of Parkinson’sdisease

Laura J Elletta,b,c David I Finkelsteinc and Victoria A.Lawsona,b,c

aThe Department of Pathology; bThe Department of Microbiologyand Immunology; cThe Florey Institute for Neuroscience andMental Health, The University of Melbourne, Victoria, Australia

CONTACT Victoria A. Lawson [email protected]

The spread of α -synuclein pathology in Parkinson’sdisease has been described as prion like with pathologythought to spread from the peripheral to central ner-vous system in at least some forms of the disease. Prionpathology and gastrointestinal dysfunction has alsobeen reported in some forms of prion disease.

FollowingMPTP-lesioning, a well characterizedmodelof Parkinson’s disease, C57BL/6 mice develop a constipa-tion phenotype characterized by reduced stool frequency.We have recently correlated this dysfunction with entericglial cell reactivity and the loss of specific neuronal popu-lations in the enteric nervous system [1]. In the presentstudy we investigated the mechanism of MPTP inducedneurodegeneration in the enteric nervous system.

MPTP induced toxicity is dependent on the uptake ofits toxic metaboliteMPP+ by dopamine active transporter(DAT) expressed on dopaminergic neurons. DAT expres-sion is highest in the ileum of the gastrointestinal tract.Consistent with this tyrosine hydroxylase (TH) immu-noreactive (IR) dopaminergic neurons were significantlyreduced in the ileum of MPTP-lesioned mice, but unaf-fected in the colon where DAT expression is lower.

Although MPTP-lesioning is not generally associatedwith Lewy pathology, MPTP-lesioning led to a changein localization of α -synuclein in DAT-IR neurons ofthe ileum. MPTP treatment did not affect α -synucleinlocalization in the colon.

Non-dopaminergic neurons identified by the expres-sion of neurofilament (NFM-IR neurons) were alsoreduced in the myenteric plexus of the ileum, but notcolon of MPTP lesioned mice.

We hypothesize that MPTP-initiated loss of non-dopa-minergic cell populations be may in response to MPTPinduced changes in α -synuclein and/or the response ofenteric glial cells to MPTP induced injury. This hypothesisis consistent with a toxic insult triggering Parkinson’s dis-ease pathology in a vulnerable cell population within thegastrointestinal tract, with secondary neurodegeneration

114 ABSTRACT

caused by the spread of α -synuclein aggregates orneuroinflammation.

Reference

[1] Ellett et al., Sci Rep. 2016;6:30269.

208. Binding prions to soils impact PrPCWD

recovery but not infectivity

Alsu Kuznetsovaa,b, Debbie McKenzieb and Judd M.Aikena

aAgricultural Life and Environmental Sciences Faculty, University ofAlberta, Edmonton, Canada; bFaculty of Science, University ofAlberta, Edmonton, Canada

CONTACT Alsu Kuznetsova [email protected]

ABSTRACT

Chronic wasting disease (CWD) is a contagious priondisease affecting cervids. Soil can act as an environmentalreservoir resulting in CWD transmission. CWD prionsshed from animals at the preclinical and clinical stages ofdisease into the environment remain infectious even afterprolonged periods. In this study, we used soil minerals(quartz, illite, montmorillonite (Mte)) as well as six surfacesoil horizons of four different soils: horizons LFH (plantlitter horizon) and Ae (illite-enriched mineral horizon) ofLuvisol, horizons LFH (plant litter) and Bf (illite and ironenriched mineral horizon) of Brunisol, and Ah (humichorizon) horizons of two Chernozemic soils. Collectedsoil samples represent soil cover of boreal, tundra(Luvisols and Brunisols) and prairie regions (Chernozems).

Long-term incubation of prions with illite, Mte andsoils results in decreased recovery of PrPCWD. Binding ofprions to minerals and soils became more avid and irre-versible with time; PrP signal on immunoblots continu-ously declined until it was no longer detectable after30 weeks incubation in soils with loamy-clay texture andMte minerology. This continual decline of PrPCWD

immunoreactivity did not correlate with prion infectivitylevels as no significant differences were found in incuba-tion periods between mice intraperitoneally inoculatedwith pure 1% brain homogenate and with the CWD-BHpre-incubated with quartz or Luvisolic Ae horizon for 1and 30 weeks. After 55 weeks incubation with Chernozemand Luvisol, bound PrPCWD was undetectable by immu-noblotting but remained infectious.

We show that prions bind to soils and remain infec-tious after extended periods of time (at least up to55 weeks). Our findings indicate that analysis of environ-mental samples for PrPCWD will be complicated as

prolonged incubation and lack of detection by westernblot does not correspond to a decrease in PrPCWD

infectivity.

209. Comparative analysis of the neuroinvasion ofChronic Wasting Disease strains (Wisc-1 and H95+)in transgenic mice expressing different cellularprion proteins

Danielle Gushuea, Camilo Duque Velásqueza, ChiyeKima, Judd Aikenb and Debbie McKenziea

aDepartment of Biological Sciences, Centre for Prions and ProteinFolding Diseases, University of Alberta, Edmonton, Canada;bDepartment of Agricultural, Food and Nutritional Sciences,Centre for Prions and Protein Folding Diseases, University ofAlberta, Edmonton, AB, Canada

CONTACT Danielle Gushue [email protected]

ABSTRACT

Chronic wasting disease (CWD) is a contagious prion dis-ease of free-ranging and captive cervids from NorthAmerica, Scandinavia, and Korea. The transmission cycleof CWD among cervids relies on the lymphotropic natureof CWDprions and the environmental persistence of prioninfectivity shed by affected animals. Multiple prion strainscirculate in CWD enzootic areas and their diversity ismodulated by disease transmission between cervids expres-sing different PrPC molecules. Prion strains neuroinvadewith different efficiency and this process is dependent onthe route of exposure, the infectious dose and the structuralcompatibility between prion conformation and host PrPC.To evaluate if the route of exposure affects strain adapta-tion, we compared the neuroinvasion properties of pureWisc-1 strain and a mixture of this one with the H95+

strain. We previously demonstrated these strains have dis-tinct biological properties, including conformational differ-ences, brain targeting areas, and transmission host range.Two transgenic mouse lines expressing PrPC allelotypes,Q95G96 or Q95S96, were exposed either orally or intraper-itoneally (IP) and the phenotypic properties produced byeach strain-route-host combination were compared.Consistent with previous observations in host expressingwt PrPC, the Wisc-1 strain had higher disease penetranceand shorter incubation periods compared to the H95+. Theintraperitoneal route wasmore efficient than the oral route,which lacked complete attack rates. The incubation periodofWisc-1 doubled compared to the intracranial (IC) route.Interestingly, the differences in incubation between the hostexposed to Wisc-1 or the H95+ mixture were smaller thanpreviously reported by IC route. The Wisc-1 host-specifictitre in theH95+mixture is at least two-fold lower than pureWisc-1, suggesting the route of exposure influenced strainselection from the mixture. This also indicates Wisc-1 is

PRION 115

highly adapted for neuroinvasion of deer expressing wt-PrPC. With the potential for new prion strains to emergeand the presence of multiple strains in a single host, it isimportant to characterize their transmission properties,including their capacity to neuroinvade and their tissuetropism.

210. Neurofilament light chain (NfL) as a possiblebiomarker for drug efficacy in mouse models ofneurodegenerative diseases

Masakazu Hirouchia,b*, Atsushi Aoyagia,b, AnnaliseBonda, Kurt Gilesa,c, Carlo Condelloa,c and Stanley B.Prusinera,c

aInstitute for Neurodegenerative Diseases, Weill Institute forNeurosciences, University of California, San Francisco, CA, USA;aDaiichi Sankyo Co., Ltd, Tokyo, Japan; cDepartment of Neurology,University of California, San Francisco, CA, USA

CONTACT Masakazu Hirouchi [email protected]

ABSTRACT

Neurofilament light chain (NfL) is known as a promisingfluid biomarker of neurodegenerative disease progres-sion. NfL changes in plasma have been found in humanswith neurodegenerative diseases (NDs) and transgenic(Tg) mouse models. Here, we report an elevation ofplasma NfL in Tg mice modelling a synucleinopathycaused bymultiple system atrophy (MSA). Brain extractsfrom deceased MSA patients caused disease in Tg(SNCA*A53T) mice expressing mutant α-synuclein(A53T). NfL levels in plasma correlated with α-synucleinprions and phosphorylated α-synuclein in the brain. Bymeasuring NfL in plasma, we were able to predict the lifespan of Tg(SNCA*A53T) mice inoculated with MSAbrain homogenates. Our findings suggest that plasmaNfL levels can be used to predict disease progression inTg mouse models of NDs. Preclinical methods for mea-suring neurodegeneration in Tg mouse models are valu-able systems for assessing efficacy in therapeutic studies.To investigate this idea further, we employed a PrPdisease model in which RML prions are used to infectFVB mice, which were treated with the aminothiazoleIND24, a previously published anti-prion compound.Plasma NfL levels increased in inoculated FVB micewithout treatment, but NfL was suppressed in micetreated with IND24, which was consistent with theirextended survival. We are currently measuring PrPSc inbrain and NfL in plasma levels in scrapie prion–infectedFVB mice to understand the relationship between drugtarget and biomarker in our study.

211. Prion infection dysregulates retromermediated trafficking

Govinda Sharma, Li Lu and Sabine Gilch

Department of Ecosystem and Public Health, Calgary PrionResearch Unit, Faculty of Veterinary Medicine, Hotchkiss BrainInstitute, University of Calgary

CONTACT Govinda Sharma [email protected])

ABSTRACT

Background: Vesicle trafficking is impaired in most ofthe neurodegenerative diseases caused by protein mis-folding. We have previously reported that membranebound Rab7 is reduced in prion infected cells. Rab7regulates the trafficking from early endosome to lateendosomes. Similarly, retromer complex mediates thetrafficking between endosomes and the Golgi appara-tus. It has been reported that the VPS35, a componentof retromer complex, is downregulated in the brains ofCreutzfeldt Jacob disease (CJD) patients. Here we stu-died how prion infection affects the retromer basedtrafficking in cultured cells and analysed the levels ofVPS35, in prion infected mice as well as cultured cells.Methods: Non-infected or prion-infected (22L-CAD5)cells were allowed to bind fluorescently labeled Choleratoxin subunit B (CTB) for 30 min at 4°C. Then the trans-port of CTB was chased at 37°C at different time pointsfollowed by immunostaining with Rab5 or Rab6 to visua-lize early endosomes or the Golgi complex, respectively.Confocal microscopy (Zeiss LSM700) and the software Zenwere used to analyse the colocalization of CTB with Rab5or Rab6. Similarly, the intracellular localization of VPS35 in22L- or non-infected Neuro2A (N2A) was studied usingimmunofluorescent staining and confocal microscopy.Prion infection was confirmed by assessing the presenceof proteinase K (PK) resistant prion protein (PrPSc) in celllysates.Western blot analysis was used to evaluate the levelsof VPS35 in non-infected or 22L-N2A cells, as well as inbrains of 22L prion- or mock (PBS)-inoculated mice125 days post intracerebral infection (dpi).Results: We observed that GM1, a sphingolipid present inthe lipid raft, is upregulated in prion-infected cells. CTBbinds toGM1 and follows the endocytic route to the Golgivia retromer-mediated transport. We found that CTBtransport from Rab5 positive early endosomes to theGolgi is delayed in prion-infected CAD5 as well as N2Acells. Also, the intracellular localization of the retromercomponent VPS35 is different in prion-infected N2A cellswhen compared to non- infected cells. It has beenreported that VPS35 levels are downregulated in CJDpatient’s brain. We found a similar decrease in VPS35levels in 22L-CAD5 cells as well as in brains of mice

116 ABSTRACT

inoculated with 22L prions when compared to mock(PBS) inoculated controls at 125 dpi.Conclusion: These findings indicate that prion infec-tion dysregulates retromer mediated trafficking, poten-tially due to the decreased VPS35 levels. Thedysregulation of intracellular vesicle trafficking couldbe a mechanism of neuronal damage caused by prioninfection and represents a novel therapeutic target.

212. Effects of E200K genetic predisposition to priondisease on cardiomyocyte cellular function,cytoskeleton structure, and protein expression

Aleksandar Wooda, Simote Foliakia, BradleyGrovemana, Jue Yuanb, Leslie Coopermanb, PaulTesarb, Wen-Quan Zoub and Cathryn Haigha

aNational Institute of Allergy and Infectious Diseases (Laboratory ofPersistent Viral Diseases); bCase Western Reserve University Schoolof Medicine

CONTACT Aleksandar Wood [email protected]

Background/Introduction: The prion protein (PrP)has been shown to have a role in cellular protectionagainst oxidative stress in numerous cell types, includ-ing murine heart tissue. Likewise, cardiac autonomicimpairment has been linked with prion disease. Ingenetic Creutzfeldt-Jakob Disease (CJD) caused by theE200K single amino acid mutation, intraneuronal PrPdeposition is found in the brainstem, which may influ-ence cardiovascular autonomic control. However, thedirect functional influence of the E200K mutation oncardiomyocytes has not been investigated. We hypothe-sized that the E200K mutation may cause a loss of PrPfunctional capacity resulting in increased oxidativedamage and deficient function.Materials and Methods: Cardiomyocytes were differ-entiated from several human induced pluripotent stemcell lines. The underlying cells were either ‘normal’ (nopredisposition to dementia), contained the E200Kmutation within the prion gene (predisposed to devel-oping genetic CJD), or trisomy 21 (causing Down’sSyndrome in the donor). Baseline electrophysiologicalrecordings were collected, measuring peak-to-peakamplitude as well as changes in overall channel activityover time. Experimental treatments were added andrecordings were continued at specified time intervals,spanning from hours to days. Treatments were used tostimulate mitochondrial function, which would beexpected to increase oxidative stress, and to inhibitcontractility, reducing workload and therefore reducingoxidative stress. Twenty-four hours post-treatment,protein expression levels were assessed via WesternBlot and Immunofluorescence.

Results: Preliminary electrophysiology recordings ofcardiomyocyte cultures revealed that contractility ofthe cells varied basally and was altered by stimulatingmitochondria. Expression levels of the superoxide dis-mutase (SOD) family was assessed as an indication ofincreased load from oxidative stressors; higher levels ofexpression can indicate compensatory efforts to negatethe increased stress. E200K cardiomyocytes revealedincreased SOD2 expression compared with controls,as well as increased prion protein (PrP) expressionand increased B-Cell Lymphoma 2 (BCL2) expression– an anti-apoptotic marker.Conclusions: The function, structure, and proteinexpression of cardiomyocytes was influenced by theE200K mutation. An impaired response to oxidativestress was indicated by changes in the localization andexpression of SODs, PrP itself, and the anti-apoptoticprotein, BCL2.

KEYWORDS: Prion; oxidative stress; reactive oxygenspecies; neurodegeneration; spongiform encephalopa-thy; cardiomyocyte; iPSC

213. Prenatal noise stress aggravates cognitivedecline and the onset and progression ofβ-amyloid pathology in a mouse model ofAlzheimer’s disease

Zahra Jafari, Megan Okuma, Hadil Karem, JogenderMehla, Bryan E. Kolb and Majid H. Mohajerani

Department of Neuroscience, Canadian Centre for BehaviouralNeuroscience (CCBN), University of Lethbridge, Lethbridge, AB,Canada

CONTACT Zahra Jafari [email protected]

ABSTRACT

Environmental distresses occurring during the sensitiveperiods of early-life may exacerbate the vulnerability todevelop physical and mental diseases in old age. Studieshave shown the impact of prenatal stress (PS) on theendocrine development and reprogramming ofhypothalamic-pituitary-adrenal (HPA) axis functionsin association with cognitive development and suscept-ibility to neuropsychiatric disease. Long-term exposureto glucocorticoids can damage the brain and intensifythe progression of Alzheimer’s disease (AD)-like neu-ropathological changes, especially in females. There is,however, less information as to the link between PS andthe risk of developing AD pathology during the life-span. Male and female APPNL-G-F/NL-G-F offspring ofdams exposed to gestational noise stress were comparedwith the control offspring in corticosterone

PRION 117

alternations, cognitive and motor performances, andthe onset age and development of amyloid beta (Aβ)plaques across age. The hyperactivity of the HPA axis,spatial learning, and Aβ development were sex-specificshowing persistent high levels of stress and furthermemory loss in females than males, especially in PSmice. The Aβ deposition was started earlier, by 2–3 months, and exhibited a heightened progression inPS animals. The PS also created a long-lasting anxiety-like behaviour and impairment in cognitive functionand motor coordination. The findings suggest PS as arisk to exacerbate AD-like neuropathological changesduring the lifespan, with higher susceptibility offemales. The findings were discussed according to themost likely mechanisms for the PS effects, i.e., dysre-gulation of the neuroendocrine system and the placentaby the PS.

KEYWORDS: Prenatal stress; Alzheimer’s disease; Aβplaque; cognitive decline; motor impairment; prepulseinhibition; HPA axis

214. Identification of novel CWD strains

Debbie McKenziea, Camilo Duque Velasqueza, AllenHerbsta, Elizabeth Triscotta, Jacques van der Merwea,Samia Hannaouib, Leonardo Corteza, Sara Amidiana,Valerie Sima, Holger Willea, Hermann Schatzlb, SabineGilchb and Judd Aikena

aCentre for Prions and Protein Folding Diseases, University ofAlberta, Edmonton, AB, Canada; bCalgary Prion Centre, Universityof Calgary, Calgary, AB, Canada

CONTACT Debbie McKenzie [email protected]

ABSTRACT

Chronic wasting disease is a set of prion diseases infect-ing captive and wild cervids. As different CWD agentsare transmitted between different cervid species andbetween the same species having different Prnp poly-morphisms, novel CWD strains can be generated.These new strains can exhibit different biochemicalproperties as well as different conformers.Identification of novel CWD strains is critically impor-tant as the different strains may vary in their hostranges, increasing the potential for transmission toeconomically important species as well as zoonotictransmission.

A rate limiting step in the characterization of CWDstrains is the identification of deer samples that poten-tially contain novel strains. Much of the strain genera-tion likely occurs during early passage betweendifferent Prnp genotypes (of the same or different spe-cies). Traditionally these differences have been

identified following passages into rodent models(strains of lab mice, transgenic mice expressing differ-ent Prnp sequences and/or hamsters). Incubation per-iods can be long and the number of potential isolates ishigh making transmission experiments a slow, tedious,and expensive process. To streamline the process, wehave identified a panel of in vitro analyses to help targetsamples for further characterization. For some isolates,potential new strains have been identified by westernblot analysis, others by cervid cell assay, changes in thePrnp sequence, folding and/or aggregation differencesas well as ability to seed reactions in RT-QuIC. Isolatesof interest are then further characterized by transmis-sion in a variety of different tg mouse lines, wild-typemice, hamsters, and voles. Using these criteria, fivepotential CWD strains have been identified.

215. Water quality improvement agent (new typeCAC-717) also asts as prion disinfectant

Takashi Onoderaa, Koichi Furusakib, Makoto Haritania

and Akikazu Sakudoc

aUniversity of Tokyo; bMineral Activation Technical Research Center;cUniversity of Ryukyus

In Prion 2018 we reported electrically charged material(CAC-717) by continuously applying an electric field tomineral water containing calcium hydrogen carbonate.A Teflon insulation-coated electrostatic field electrode(N-800N, Mineral Activation Technical ResearchCenter; Japan Patent No. 5864010) was used to createthe electric field at a voltage of 2 × 104 V for 48 h.CAC-717 solution in distilled water (Japan Patent No.5778328, FDA/USA Regulation No. 880.6890 Class 1disinfectant) had a pH of 12.39 ± 0.03 and containedcalcium hydrogen carbonate particles (1,120 mg/L) andcarbonate complex microparticles (50–500 nm) with amesoscopic structure. CAC-717 works as a soil amend-ment was evaluated by growth of green tea leaves inJapanese farm lands. In Prion2019 we report that CAC-717 absorbed by ceramics were applied to fresh or seawater to work as water quality improvement agents.

Mouse prion (Chandler) infected N2a cell (ScN2a)lysate was subsequently mixed with equal volume ofCAC-717 solution. As a negative control, uninfectedN2a cell lysate was diluted 2-fold with PBS or RIPAbuffer. 200 μg lysate protein was diluted in 100 μg ofbuffer (PBS or RIPA) and mixed with 100 μℓ of PBS.Mixtures were incubated at room temperature for 1 hthen immediately diluted in PBS before determiningthe amount of prion in each sample for Western blot-ting with SAF83 monoclonal antibody. Prion was notdetected in the ScN2a cell line lysate mixed with CAC-

118 ABSTRACT

717 solution after testing with Western Blotting.However, PrPC levels was intact in both uninfectedN2a cell and infected ScN2a cells, detected byWestern Blotting.

CAC-717 ceramics placed inside concrete blockswere sprayed into sea water by the seashore ofShimane Prefecture, Japan. Then alteration of waterenvironment was observed for 6 months. Number ofsea urchin, sea cucumber, and abalones were calculatedinside the concrete blocks with CAC-717 ceramics.Yield, as calculated by the number of these marineanimals, was then determined. No animal was observedin the blocks with ceramics without CAC-717. Whilelarge amounts of animals were attracted and growinginside the blocks with CAC-717 ceramics.

Evaluation of water environment, nutrient content,composition, and microbiological activity tend toincrease following use of water quality improvementagents. Numerous studies have therefore to be exam-ined for the effect of water quality improvement agent,beside animal yield and quality. Our future aims are todetermine the microbiological activity of CAC-717ceramics after spraying with CAC-717.

216. Molecular dynamics study of PrP conversion

Lyudmyla Dorosha,b, Min Wua, SandipanChakrabortyc, Holger Willed,e and Maria Stepanovaa,b

aDepartment of Electrical and Computer Engineering, University ofAlberta, Edmonton, Canada; bNational Institute for NanotechnologyNRC; Edmonton, Canada; cDepartment of Microbiology, Universityof Calcutta, Kolkata, India; dDepartment of Biochemistry, Universityof Alberta, Edmonton, Canada; eCentre for Prions and ProteinFolding Diseases, University of Alberta, Edmonton, Canada

CONTACT Lyudmyla Dorosh [email protected]

ABSTRACT

Prion diseases are believed to be caused by a conversionof native cellular prion protein (PrPC) into infectious β-rich aggregates (PrPSc). The structures of these aggre-gates and the molecular mechanisms of the conversionremain unknown, although numerous models for thestructure of PrPSc have been suggested [1]. In order toelucidate mechanisms of the conversion, we have doneextensive all-atom molecular dynamics (MD) simula-tions tackling early stages of this process.

We employed several β-helical models of PrPSc.These include the recently proposed [2] 4-rung right-handed β-helical (RHBH) head-to-tail moPrPSc(89–230) model based on cryo-EM and X-ray fiber diffrac-tion data and corrected with MD simulations; hypothe-tical 4-rung RHBH head-to-head and head-to-tailmoPrPSc(89–230) models [3]; and a 4.5 rung left-

handed β-helical (LHBH) head-to-head huPrPSc(114–220) model built by computational threading of the PrPsequence onto published β-solenoid architectures. Insimulations of the PrPC conversion, matching nativePrPC constructs (solution NMR) were also included.

In our MD simulations of the above-mentionedPrPSc models, the head-to-tail RHBH dimer [2]showed a higher stability in comparison to otherconstructs. However in different dimeric models, apreference for head-to-head configurations wasfound. A greater structural stability and an increaseof β-content were also found in the dimers in com-parison to monomers. The β-content and morphol-ogy were somewhat less stable in the LHBH modelthan in the RHBH models.

Simulations of the PrPC conversion containingnative PrPC and PrPSc revealed early unfolding eventsin PrPC. Head-to-head contact of PrPC with a RHBHPrPSc resulted in unfolding and formation of β-strandsdistant from the contact area, in regions of loop LH2H3and helices H2 and H3, in first 100 ns. These changesremained stable for the next 150 ns. In systems withtwo PrPC molecules, similar trends towards partialunfolding of H2 and H3 with formation of β-strandswere observed. During 155 ns simulations of nativePrPC in a head-to-tail contact with monomeric RHBHPrPSc [3], a pronouncedly better connection wasobserved. Distinct from other systems, the S1S2 bundleof PrPC approached the β-solenoid, partial unfolding ofhelix H1 occurred, and some of secondary structurearound the loop LH2H3 was lost. However, our resultssuggest that fast conversion of PrPC has a low prob-ability. The connection between S1, S2, and H2H3bundle is not interrupted, stabilizing the remainingsecondary structure in PrPC.

References

[1] Requena, Wille. Prion. 2014;8:60–66.[2] Spagnolli et al., bioRxiv 2018; Preprint http://dx.doi.

org/10.1101/505271.[3] Requena, Chakraborty, personal communication.

217. The presence of autoantibodies against theprion protein is independent of PRNP mutations

Karl Frontzeka, Manfredi Cartaa, Marco Losaa, MirkaEpskampa, Georg Meislb, Ulrike Camenischc, TuomasKnowlesb, Simone Hornemanna and Adriano Aguzzia;the THAUTAN-MC InvestigatorsaInstitute of Neuropathology, University of Zurich, Zurich,Switzerland; bDepartment of Chemistry, University of Cambridge,Cambridge, UK; cInstitute of Surgical Pathology, University ofZurich, Zurich, Switzerland

PRION 119

CONTACT Karl Frontzek [email protected]

ABSTRACT

Background: Prion diseases are incurable diseases ofthe central nervous system, which not only occur assporadic and infectious forms, but can also be trans-mitted through the germ line as autosomal dominanttraits. What is more, genetic prion diseases usuallymanifest in late age, indicative of protective factors.Here we reasoned that subtle conformational altera-tions of pathogenic prion protein (PrPC) variantscould stochastically generate immunogenic neo-epi-topes, raising the question whether such protectivefactors may consist, at least in part, of anti-PrPC auto-antibodies. We therefore conducted an extensive searchfor such autoantibodies in persons carrying pathogenicPRNP mutations and, for control, in their unaffectedrelatives.Methods: In this case-control study, we have collectedblood samples from individuals (n = 134) expressing awide spectrum of disease-associated variants of thePRNP gene. Individuals with a positive family historyof genetic prion disease (n = 79) but lacking disease-associated PRNP mutations served as controls.Antibody reactivity was measured using an indirectELISA for the detection of human IgG1-4 antibodiesagainst full-length human prion protein. We evaluatedthe effects of age, gender, PRNP mutations, PRNPcodon 129 polymorphism, clinical signs of prion dis-ease, and total IgG levels on autoantibody reactivity. Ina subset of patients, autoantibody reactivity wasassessed longitudinally for up to 2 years after baselinemeasurements.Results: We found that antibody reactivity was present ina subset of both PRNP mutation carriers and controls.Elder individuals were at lower odds for the presence ofanti-PrPC autoantibodies, independently of total IgGlevels or PRNP genotype. Gender, clinical signs of priondisease, pathogenic PRNP variants or p. 129 polymorph-ism did not influence autoantibody reactivity. Moreover,levels of anti-PrPC IgG were stable over up to 2 years.Conclusions: This study shows that pathogenic PRNPvariants do not notably stimulate antibody-mediatedanti-PrPC immunity. Moreover, anti-PrPC IgG autoan-tibodies are independent of overt signs of prion disease.The presence of anti-PrPC autoantibodies in the generalpopulation without any disease-specific associationsuggests that relatively high titers of naturally occurringantibodies are tolerated. This observation suggests thatsuch antibodies, if administered to humans, may notinduce severe adverse reactions and may therefore serveas anti-prion therapeutics.

218. Single-molecule studies show α-synucleinaggregation is inhibited by anti-prion compounds andnovel computationally-derived ligands

Chunhua Dongaa, Lindsay Sheareraa, Craig R.Garenaa, Pascal Mercierbb, Nils O. Petersencc, andMichael T. Woodsideaa

aDepartment of Physics, University of Alberta, Edmonton, AB,Canada; bNANUC National High Field Nuclear Magnetic ResonanceCentre, Edmonton, AB, Canada; cDepartment of Chemistry,University of Alberta, Edmonton, AB, Canada; Edmonton, AB,Canada

ABSTRACT

The intrinsically disordered protein α-synuclein (αS) isenriched in neurons and has been shown to be linkedto Parkinson’s disease (PD) and other related neurode-generative disorders. Single αS molecules are postulatedto undergo a misfolding/aggregation cascade that ulti-mately yields Lewy bodies, the histopathological hall-mark of PD. The main neurotoxic species in thisprocess have been shown experimentally to be pre-fibrillar oligomeric intermediates. We have investigatedthe inhibitory effects of various ligands on αS aggrega-tion using single-molecule fluorescence cross correla-tion spectroscopy techniques and traditional ensembleThT assays. These methods monitor respectively theinfluence of the ligands on the nucleation of smalloligomers and the formation of fibrils. We character-ized several inhibitors of αS aggregation from amongknown anti-prion agents and from a small library ofcompounds that were predicted to inhibit dimer for-mation based on computer simulations of αS dimerstructures elucidated using single molecule force spec-troscopy measurements of dimer aggregation. The abil-ity of the same ligands to inhibit PrP aggregation andαS aggregation suggests there may be common featuresin the aggregation mechanisms that can be targeted forinhibition. The success of novel aggregation inhibitorsdiscovered through computational simulations suggeststhat this approach provides a viable way to search fornew inhibitors. Looking forward, we will broaden oursearch for additional novel candidates for developmentof PD therapeutics by testing αS with known inhibitorsof other proteins that form amyloids (Aβ, tau, hunting-tin, prion etc.), and by expanding the in silico discoveryplatform.

References

[1] Spillantini et al. PNAS USA. 1998;95:6469-73.[2] Masliah et al. Science. 2000;287:1265-69.[3] Volles et al. Biochem. 2003;42:7871-8.

120 ABSTRACT

[4] Schwille et al. Biophys. J. 1997;72:1878–86.[5] Saeed. Et al. Am. J. Clin. Pathol. 1967;309:274-84.[6] Priola et al. Science 2000;287:1503-6.[7] Dee et al. Biochim. Biophys. Acta. 2012;1824:826-32.

219. Domain-specific quantification of PrP incerebrospinal fluid by targeted mass spectrometry

Eric Vallabh Minikela,b,c, Eric Kuhnd, Alexandra Coccod,Sonia M Vallabha,b,c, Chrissy Hartigand, Andrew GReidenbacha, Franc Llorense,f, Inga Zerre, SabinaCapellarig,h, Piero Parchig,i, Stuart L. Schreibera,j,Steven A. Carrd

aCenter for the Science of Therapeutics, Broad Institute of MIT andHarvard, Cambridge, MA, USA; bProgram in Biological andBiomedical Sciences, Harvard Medical School, Boston, MA, USA;cPrion Alliance, Cambridge, MA, USA; dProteomics Platform, BroadInstitute of MIT and Harvard, Cambridge, MA, USA; eNationalReference Center for TSE, Georg-August University, Göttingen,Germany; fBiomedical Research Networking Center onNeurodegenerative Diseases (CIBERNED), L’Hospitalet deLlobregat, Barcelona, NA, Spain; gIRCCS – Institute of NeurologicalSciences, Bologna, Italy; hDepartment of Biomedical andNeuromotor Sciences, University of Bologna, Bologna, Italy;iDepartment of Experimental, Diagnostic and Specialty Medicine,University of Bologna, Bologna, Italy; jDepartment of Chemistry &Chemical Biology, Harvard University, Cambridge, MA, USA

CONTACT Eric Vallabh Minikel [email protected]

ABSTRACT

Antisense oligonucleotides in preclinical developmentfor prion disease will seek to lower PrP expression inthe brain [1]. Trials of PrP-lowering therapies arelikely to rely on quantification of PrP in cerebrosp-inal fluid (CSF) as a pharmacodynamic biomarkerand possibly as a trial endpoint [1,2]. Studies usingPrP ELISA kits have reproducibly shown that CSFPrP is lowered in the symptomatic phase of disease[2–6], a potential confounder for reading out theeffect of PrP-lowering drugs in symptomatic patients.To date it has been unclear PrP’s lowered abundancein CSF results from its incorporation into plaques[7], retention in intracellular compartments [8],downregulation as a function of the disease process[9], or other factors. Because misfolding or proteoly-tic cleavage could potentially render PrP invisible toELISA even if its concentration were constant orincreasing in disease, we sought to establish anorthogonal method for CSF PrP quantification. Wedeveloped a targeted mass spectrometry methodbased on multiple reaction monitoring (MRM) [10]of nine PrP tryptic peptides quantified relative toknown concentrations of isotopically labelled stan-dards. Analytical validation experiments showedtechnical replicate coefficients of variation below

15%, strong dilution linearity and spike recovery,and applicability to both CSF and brain homogenateand across humans as well as preclinical species ofinterest. In N = 55 CSF samples from individualsreferred to prion surveillance centres with rapidlyprogressive dementia, all six human PrP peptides,spanning the N- and C-terminal domains of PrP,were uniformly reduced in prion disease cases com-pared to non-prion diagnoses, replicating the find-ings from ELISA studies. Our study providesevidence that lowered CSF PrP concentration inprion disease is a genuine result of the disease pro-cess and not merely an artefact of ELISA-based mea-surement, and we provide a novel method suitablefor preclinical and clinical quantification of CSF PrPas a tool for drug development.

References

[1] Vallabh, et al., In preparation.[2] Vallabh, et al., bioRxiv 2018. DOI:10.1101/295063.[3] Meyne, et al., J Alzheimers Dis. 2009;17:863.[4] Dorey, et al., JAMA Neurol. 2015;72:267.[5] Abu Rumeileh et al, J Alzheimers Dis. 2017;55:1471.[6] Villar-Pique, Schmitz et al., Mol Neurobiol. 2018.

DOI:10.1007/s12035-018–1251-1.[7] Parchi et al., Ann Neurol. 1999;46:224[8] Goold, et al., J Cell Sci. 2013, 126, 3552.[9] Mays, et al., J Clin Invest. 2014;124:847.[10] Carr, et al., Mol Cell Proteomics. 2014;13:907.

220. Light and electron microscopic studies of the139A-H(Ha) strain of scrapie passaged in hamsters

Pawel P. Liberski a, Agata Gajos b, Beata Sikorska a

and Janusz Moryś c

aLaboratory of Electron Microscopy and Neuropathology,Department of Molecular Pathology and Neuropathology, Chair ofOncology, Medical University Lodz, Poland; bDepartment ofExtrapyramidal Diseases, Chair of Rehabilitation, MedicalUniversity of Lodz, Lodz, Poland; cDepartment of Anatomy andNeurobiology, Medical University of Gdansk, Gdansk, Poland

ABSTRACT

We report here the light and electron microscopicneuropathology of the 139A-H strain of scrapie pas-saged in Syrian golden hamsters.Material and methods: This strain was derived fromthe Scrapie Sheep Brain Pool (SSBP)-1 lineage [3,5].Briefly, scrapie from a natural source was passagedthrough Cheviot sheep, Welsh Mountain sheep,goats, mice and led to the isolation of the‘Chandler’ scrapie strain. The clone 139A strain wasderived following passage in hamsters and this strain

PRION 121

was made available for us by Dr. Richard I Carp,IBR, Staten Island, USA.Analysis of PrP immunostaining: The intensity of theimmunostaining were quantified in a single hamsterbrain using the images obtained from the AxioScan.Z1 system (Zeiss, Germany) using the Zeiss Zen 2.3(Blue Edition) Software.Results: The general neuropathological picture con-sisted of spongiform change and severe astrocytic glio-sis. The topography of PrP was variable, the highestsignal was observed in the CA2-molecular layer, CA1-pyramidal and entorhinal cortex. Double immunofluor-escence stainings showed dot-like immunoreactivity ofmicrotubule associated protein (LC3) in the cell bodieswhile axons more often showed fibrillar pattern.Interestingly, some neuronal processes showed onlyneurofilament protein and LC3 positive structureswere depleted of NFP immunoreactivity. The electronmicroscopy consisted of:

(1) Spongiform vacuoles – these are always mem-brane bound and contained secondary vacuoles(i.e. membrane-bound compartments or vesi-cles within vacuoles) and curled membranedfragments.

(2) Tubulovesicular structures (TVS) – these arevesicular structures of approximate 27 nm indiameter within neuronal processes – i.e. axonalterminal or dendrites. TVS are smaller and ofhigher electron density than synaptic vesicles.The significance of TVS remains unknown.

(3) Dystrophic neurites. Dendrites or axonal preterm-inals and terminals filled with electron-densebodies, including small autophagic vacuoles.

(4) Apoptotic cell nuclei.(5) ‘Whorls’, a concentric arrays of membranes

were visible. A significance of those structuresis unknown.

PPL and BS are supported by National Science CentrePoland, grant number UMO-2015/19/B/NZ4/03234

KEYWORDS: Scrapie; electron microscopy; topogra-phy of lesion

221. Purification of small, non-fibrillar andinfectious prion particles from the brain of patientswith GSS-A117V

Ilaria Vannia, Laura Pirisinua, Claudia Y. Acevedo-Morantesb, Michele A. Di Baria, Claudia D’Agostinoa,Stefano Marcona, Geraldina Riccardia, Elena Espositoa,

Pierluigi Gambettic, Umberto Agrimia, Holger Willeb

and Romolo Nonnoa

aIstituto Superiore di Sanità, Department of Food Safety, Nutritionand Veterinary Public Health, Rome, Italy; bCentre for Prions andProtein Folding Diseases & Department of Biochemistry, Universityof Alberta, Edmonton, Alberta, Canada; cDepartment of Pathology,Case Western Reserve University, Cleveland, Ohio, USA

CONTACT Ilaria Vanni [email protected]

ABSTRACT

Gerstmann-Sträussler-Scheinker disease (GSS) is an inher-ited prion disease associated with mutations in the PRNPgene. We recently reported that GSS with the A117Vmutation, characterized by PrPSc with a protease resistantcore made up of N- and C-terminally cleaved PrP peptidesof ̴7 kDa (7 kDa PrPres), is transmissible in bank voles.However, definite proof of the infectious nature of thistruncated PrPSc type is still missing. We thus attemptedto evaluate the GSS-A117V 7 kDa PrPres role in terms ofinfectivity, strain characteristics, and structural features.

At first, we determined if the infectivity of GSS-A117VPrPSc is resistant to PK, by parallel end-point titrations involes of PK-treated or untreated brain homogenates.Unexpectedly, GSS-A117V showed an extremely highinfectious titer (109.3 ID50 U/g), which was not signifi-cantly affected by PK treatment (109.0 ID50 U/g). We thenaimed at purifying the PK-resistant GSS-A117V prions byusing a recently published protocol1 in order to determinethe amount of infectivity and PrPres in the different frac-tions, alongside with the morphological characteristics ofpurified PrPres aggregates by EM.

Purified pellet fractions from GSS-A117V containedthe expected N- and C-terminally cleaved 7 kDa PrPres,although the yield of PrPres was lower than that of ascrapie sample used as a control. We found that thislower yield depended on the low density/small size ofGSS-A117V PrPres, as it was mainly retained in the lastsupernatant fraction. All fractions were highly infec-tious, thus confirming the infectious nature of the 7kDa PrPres, with infectivity levels that directly corre-lated with the PrPres amount detected. Finally, EManalysis of these fractions showed no presence of amy-loid fibrils, only very small and indistinct, non-fibrillarPrPSc particles were detected. In contrast, PrPSc fibrilswith the expected morphology were recovered from thescrapie control sample.

Our study demonstrates that purified aggregates of 7kDa PrPres are fully infectious and encode the biochem-ical and biological strain features of the original sample,implying that the C-terminus of PrPSc is dispensable forinfectivity and strain features. Finally, the non-fibrillarmorphology of these unglycosyilated and non-GPI-anchored, small and indistinct prion particles, coupled

122 ABSTRACT

to the very high levels of infectivity detected in GSS-A117V, is reminiscent of previous data on non-fibrillarprion particles as the most efficient initiators of TSEdiseases [2]. Our findings provide new molecular andmorphological constraints on the structure of infectiousprions.

References

[1] Wenborn, et al., 2015.[2] Silveira, et al., 2005.

222. The impact of cellular prion proteinexpression levels in 5xFAD mouse behaviour andlifespan

Angela Silva-Correiaa, Matthias Schmitzb and IngaZerrc

aMedical University of Göttingen; bGeorg-August University;cNational TSE Reference Center, Göttingen, Germany

ABSTRACT

Alzheimer’s disease is the most prevalent dementiacausing progressive impairment of cognition, mood,speech, task performance ability, behaviour, andmemory. Since all treatments for AD are primarilysymptomatic, the development of disease modifyingapproaches continues to be an area of intense focus.

Previous studies already indicated an important role ofthe cellular prion protein (PrPC) as a regulator of the A βmediated toxicity by acting as a potential receptor. In thisproject we intended to evaluate different expression levelsof PrPC and its impact of amyloid pathology in ouranimal model. We had already established three differentbi-transgenic mouse line 5xFADPrnp+/+, 5xFADPrnp±

and 5xFADPrnp−/− by crossing of PrPC knockout(Prnp−/−) mice with 5xFADPrnp+/+ mice (Tg6799) exhi-biting mutations in the APP and presenilin genes. Weobserved a direct influence of PrPC expression levels on Aβ -induced behaviour deficits in our mice lines.5xFADPrnp−/− mice showed the biggest delay of beha-viour impairment (cognition, anxiety or nest building)and 5xFADPrnp+/+ mice showed the fastest onset ofbehaviour impairment. Since a PrPC-knockout cannotprevent A β -induced deficits, we propose that furtherproteins and co-factor are involved in the uptake of A βand the accompanied cellular dysfunctions.

223. Scrapie in white-tailed deer: a strain of theCWD agent that efficiently transmits to sheep?

Justin J. Greenleea, Robyn D. Kokemullera, S. JoMoorea and Heather West Greenleeb

aVirus and Prion Research Unit, National Animal Disease Center,USDA, Agricultural Research Service, Ames, IA, USA; bDepartment ofBiomedical Sciences, Iowa State University College of VeterinaryMedicine, Ames, IA, USA

CONTACT Justin J. Greenlee [email protected]

ABSTRACT

Scrapie is a transmissible spongiform encephalopathyof sheep and goats that is associated with widespreadaccumulation of abnormal prion protein (PrPSc) in thecentral nervous and lymphoid tissues. Chronic wastingdisease (CWD) is the natural prion disease of cervidspecies, and the tissue distribution of PrPSc in affectedcervids is similar to scrapie in sheep. There are severallines of evidence that suggest that multiple strains ofCWD exist, which may affect the agent’s potential totransmit to hosts of the same or different species. Weinoculated white-tailed deer with the scrapie agent fromARQ/ARQ sheep, which resulted in 100% attack ratesby either the intracranial or oronasal route of inocula-tion. When examining tissues from the brainstems orlymphoid tissues by traditional diagnostic methodssuch as immunohistochemistry or western blots, it isdifficult to differentiate tissues from deer infected withscrapie from those infected with CWD. However, thereare several important differences between tissues fromscrapie-infected white-tailed deer (WTD scrapie) andthose infected with CWD (WTD CWD). First, there aredifferent patterns of PrPSc deposition in the brains ofinfected deer: brain tissues from deer with WTD scra-pie had predominantly particulate and stellate immu-noreactivity whereas those from deer with WTD-CWDhad large aggregates and plaque-like deposits. Secondly,the incubation periods of WTD scrapie isolates arelonger than CWD isolates in mice expressing cervidprion protein. Most notably, the transmission potentialof these two isolates back to sheep is distinctly different.Attempts to transmit various CWD isolates to sheep bythe oral or oronasal routes have been unsuccessfuldespite observation periods of up to 7 years. However,WTD scrapie efficiently transmitted back to sheep bythe oronasal route. Upon transmission back to sheep,the WTD scrapie isolate exhibited different phenotypicproperties when compared to the sheep receiving theoriginal sheep scrapie inoculum including differentgenotype susceptibilities, distinct PrPSc deposition pat-terns, and much more rapid incubation periods intransgenic mice expressing the ovine prion protein.The scrapie agent readily transmits between sheep anddeer after oronasal exposure. This could confound theidentification of CWD strains in deer and the eradica-tion of scrapie from sheep.

PRION 123

224. NMR fragment screening to discover lowmolecular weight prion protein binders

Andrew Reidenbach, Eric Minikel, Sonia Vallabh,Alison Leed, Jenna Yehl, Jayme Dahlin, FlorenceWagner, Chris Lemke, Colin Garvie, Virendar Kaushik,Mike Mesleh and Stuart Schreiber

Broad Institute of MIT and Harvard

CONTACT Andrew Reidenbach [email protected]

ABSTRACT

Prion disease is a rapidly progressive neurodegenera-tive disorder caused by misfolding and aggregation ofthe prion protein (PrPC), and there are currently notherapeutic options. Attempts to target and disruptmisfolded PrP (PrPSc) have resulted in emergent drugresistance and molecules that lack efficacy on humanprion strains. Ligands of PrPC could antagonize prionformation by stabilizing the native protein or bytargeting it for degradation, but no validated small-molecule binders have been discovered to date. Wescreened 6631 low molecular weight fragments forrecombinant human PrP binding using 19F andsaturation transfer difference (STD) NMR. 80 activeswere re-tested using protein-observed transverserelaxation optimized spectroscopy (TROSY), anorthogonal 2D NMR method. A single benzimidazolederivative validated in concentration-response and itsanalogues exhibited a structure-activity relationship,albeit with weak affinity (Kd >1 mM). The exception-ally low hit rate and weak affinity observed heresuggest that PrP is a difficult target for small mole-cule binders. Nevertheless, our studies may provide astarting point for structural biology and medicinalchemistry efforts to identify more potent ligandsthrough structure-based drug design.

KEYWORDS: Fragment screening; NMR; binders

225. Public acceptability of chronic wastingdisease management approaches in Canada: apaired comparison approach

Geoffrey Durocher, Marty (M.K.) Luckert, EllenGoddard and Vic (W.L.) Adamowicz

Department of Resource Economics and Environmental Sociology,University of Alberta, Edmonton, Alberta, Canada

CONTACT Geoffrey Durocher [email protected]

ABSTRACT

Background: Chronic Wasting Disease (CWD) is aprion disease found among both captive and wildpopulations of cervids and its prevalence has beenincreasing over time. As CWD spreads, a number ofstakeholders are affected, including hunters, land-owners, and the general public. When deciding uponmanagement options for CWD, it is important to con-sider the preferences of the stakeholders involved. Thegoal of this research is to investigate the acceptability ofCWD management approaches through the eyes ofstakeholders.Methods: We employ a paired comparison approach,where respondents indicate their preferences across aseries of proposed pairs of CWD management options.Management options were developed based on prac-tices that have been used in CWD-endemic areas andbased on ideas expressed by focus groups that we con-ducted with decision-makers representing various sta-keholder interests. We develop separate series ofmanagement options for the general public, hunters,and landowners. Responses were collected fromapproximately 5200 individuals across Canada usingan internet-based survey and a sample from an onlinepanel. Respondents were asked to choose between pairsof management options, while considering their prefer-ences associated with the alternative approaches. Aftereach pair of options, respondents were asked whetherthey preferred their chosen option, or no action betaken. Preferences were analysed using conditionallogistic regressions-based on random utility models.Results: Little heterogeneity existed in the preferencesof different stakeholders from different demographicgroups. For the most part, reducing deer numbers inhigh risk CWD areas through the increased use ofhunters was preferred to using government sharpshoo-ters. Likewise, restricting the movement of carcassesand hunted products was also generally positivelyreceived by most groups, with the notable exceptionof hunters. There were also options that were almostuniversally disliked. For example, increasing incentivesfor private landowners to allow hunting on their lands,by allowing them to charge hunters for access, was evendisliked by landowners. We also find that most respon-dents prefer some action to inaction.Conclusions: As CWD increases in spread and pre-valence, policymakers are deciding which manage-ment options should be pursued. Our results providepolicymakers with important information regarding

124 ABSTRACT

public preferences for some management options thatare available for CWD. When formulating manage-ment plans, consideration of these preferences couldhelp establish and maintain public support for addres-sing CWD. With changing technology regarding themonitoring of CWD, new options are emerging thatwill warrant further inquiries into public preferences.

KEYWORDS: Chronic wasting disease; preferences;paired comparison

226. Efficient RT-QuIC seeding activity for α-synuclein in olfactory mucosa samples of patientswith Parkinson’s disease and Multiple SystemAtrophy

Chiara Maria Giulia De Lucaa, Antonio Emanuele Eliab,Sara Maria Portaleonec, Federico Angelo Cazzanigaa,Martina Rossid, Edoardo Bistaffaa, Elena De Ceccod,Joanna Narkiewiczd, Giulia Salzanod, Olga Carlettaa,Luigi Romitob, Grazia Devigilib, Paola Soliverib, PietroTiraboschia, Giuseppe Legnamed, Fabrizio Tagliavinie,Roberto Eleoprab, Giorgio Giacconea and FabioModaa

aUnit of Neurology 5 and Neuropathology, Fondazione IRCCS IstitutoNeurologico Carlo Besta, Milano, Italy; bUnit of Neurology I – Parkinsonand Movement Disorders Unit, Fondazione IRCCS Istituto NeurologicoCarlo Besta, –Milano, Italy; cDepartment of Health Sciences, Universitàdegli Studi di Milano, Otolaryngology Unit, San Paolo Hospital, Milano,Italy; dDepartment of Neuroscience, Scuola Internazionale Superiore diStudi Avanzati (SISSA), Laboratory of Prion Biology, Trieste, Italy;eScientific Directorate, Fondazione IRCCS Istituto Neurologico CarloBesta, Milano, Italy.

CONTACT Fabio Moda [email protected]

ABSTRACT

Parkinson’s disease (PD) is a neurodegenerative disorderwhose diagnosis is often challenging because symptomsmay overlap with neurodegenerative parkinsonisms. PDis characterized by intraneuronal accumulation of abnor-mal α-synuclein in brainstem while neurodegenerativeparkinsonisms might be associated with accumulation ofeither α-synuclein, as in the case of Multiple SystemAtrophy (MSA) or tau, as in the case of CorticobasalDegeneration (CBD) and Progressive Supranuclear Palsy(PSP), in other disease-specific brain regions. Their defi-nite diagnosis can be formulated only neuropathologi-cally by detection and localization of these aggregates,considered disease-specific biomarkers (DSB), in thebrain. Compelling evidence shows that trace amount ofDSB can appear in peripheral tissues, including receptorneurons of the olfactory mucosa (OM), but their con-centration is well below the limits of detection of theconventional diagnostic techniques.

We have therefore exploited the ultrasensitive RealTime Quaking Induced Conversion (RT-QuIC) assayfor the analysis of OM collected from patients with aclinical diagnosis of PD, MSA, CBD and PSP. Ourresults showed that PD and MSA samples inducedstronger RT-QuIC seeding activity for α-synucleincompared to CBD and PSP samples. Notably, the finalRT-QuIC aggregates obtained from MSA and PD sam-ples demonstrated peculiar biochemical and morpholo-gical features useful for their discrimination. Theseresults suggest that RT-QuIC represents an importanttool for the diagnostic workup for PD and neurodegen-erative parkinsonisms

KEYWORDS: RT-QuIC; olfactory mucosa; α-synu-clein; Parkinson’s Disease; parkinsonisms

227. Investigating the effect of familial mutations intau on the conformation and biological activityof tauprions

Victor Banerjeea, Gregory E. Mertza, Eric Tsea,b, Nick A.Parasa,c, Amanda L. Woermana,c, Daniel R.Southwortha,b,and Stanley B. Prusinera,b,c

aInstitute for Neurodegenerative Diseases, Weill Institute forNeurosciences, University of California, San Francisco, San 11865Francisco, CA, USA; bDepartment of Biochemistry and Biophysics,University of California, San Francisco, San Francisco, CA, USA;cDepartment of Neurology, University of California, San Francisco,San Francisco, CA, USA

Abstract not available.

228. Cervid Prnp polymorphism at codon 116generates new and distinct CWD strains

Samia Hannaouia, Elizabeth Triscottb, Camilo DuqueVelásquezb, Debbie McKenzieb, Sabine Gilcha

aDepartment of Ecosystem and Public Health, Calgary PrionResearch Unit, Faculty of Veterinary Medicine; Hotchkiss BrainInstitute; University of Calgary, Calgary, Alberta, Canada; bCenterfor Prions and Protein Folding Diseases, University of Alberta,Edmonton, Alberta, Canada

ABSTRACT

Chronic wasting disease (CWD) is a prion disease of cer-vids. To assess the role of strain emergence and adaptationin CWD, we have characterized the impact of the 116(A >G) polymorphism of the white-tailed deer (WTD)prion protein on CWD infection. Previously, we comparedCWD prion isolates from WTD encoding wild-type(116AA) or polymorphic (116AG) PrPC. We found that116AG-prions were conformationally less stable and moresensitive to proteases; infection of primary neuronal cul-tures with these isolates showed reduced infectivity

PRION 125

associated with the WTD-116AG isolate and a lower seed-ing activity in a cell-free conversion assay. Moleculardynamics simulations suggested that the structure of116G-PrPC was more flexible than 116A-PrPC.

To understand the transmission and strain properties ofthe WTD-116AG isolate, we serially passaged it and theWTD-116AA (Wisc-1) prions in tg(CerPrP)1536+/+ miceoverexpressing deer 116AA-PrPC. Upon the first passage,the 116AG isolate had a longer incubation period than theWisc-1 isolate; interestingly, the differences in biochemicalproperties between Wisc-1 and WTD- 116AG wereretained supporting the characterization of at least onenew prion strain amongst the 116AG-prions.

Upon secondary passage, infectivity of 116AG-prionswas significantly enhanced in 80% of the mice and theconformational features were retained. Remarkably, thispassage allowed us to distinguish two populations of micewith a short and a long incubation period. Although onfirst passage, only one homogenous population wasobvious, subsequent passages allowed a better distinctionand characterization of two 116AG-prions (short and longincubation). Biological cloning in tg(CerPrP)1536+/+ micethrough inoculation of 10-fold serial dilutions of the116AA and 116AG deer brain homogenates confirmedthe presence of a second strain in the 116AG isolate, yetthe slow disease progression feature of the polymorphicisolate was still striking for both of the 116AG-prions(short and long incubation) compared to the rapid diseaseprogression of the 116AA. Interestingly, upon deglycosyla-tion, PK-resistant fragments with different electrophoreticmobility are observed. Further confirmation of two strainspresent in 116AG was obtained following passage of thisinocula in SyrianGoldenHamsters. Ten of twelve hamstersinfected with 116AG were positive for PrPres. Western blotanalysis of PrPres showed two different migration patterns,high molecular weight (7/10) and low molecular weightmigration pattern (3/10). Our data support the hypothesisthat two strains co-existed, as dominant and minor strains,in the 116AG deer.

Our findings give insight into the variability of CWDstrains, which can have important implications in termsof their possible distinct capacities to cross speciesbarriers into both cervid and non-cervids.

229. Aberrations of Amyloid-β in slow and rapidlyprogressive dementias

Aneeqa Noora*, Saima Zafara*, Hassan Dihazib andInga Zerra

aPrion Research Group, National Reference Center for TSESurveillance, Department of Neurology, Georg-August University,Göttingen, Germany; bDepartment of Nephrology andRheumatology, Georg-August University, Göttingen, Germany

*These authors contributed equally to this work

CONTACT Aneeqa Noor [email protected]

ABSTRACT

Amyloid-β (Aβ), one of the key players in Alzheimer’sdisease (AD), has the capability to exist as various proteo-forms [1]. Over the last decade the self-seeding capabilityand relative pathogenicity of these proteoforms has beenan active target of research however, their distinct clinicalinvolvement in neurodegeneration is yet to be elucidated.For this purpose, we aimed to decipher the prion-likestructural and functional aspects of Aβ aggregation inrapidly progressive dementias.

We used affinity-based proteomic profiling to isolateAβ from brains of AD, rapidly progressive AD (rpAD)and non-demented controls and established a signature ofbrain-derived proteoforms, adding to the evidence thatAβ40 and Aβ42 are not sole players in progression of AD.Our preliminary experiments reveal a differential involve-ment of 26 different proteoforms in AD and rpAD brain.Quantification of various Aβ cleaving enzymes in thesame samples supported the generation of distinct proteo-forms in AD and rpAD. To further understand the patho-logical significance of Aβ in slow and rapidly progressivedementias, we isolated interacting partners of Aβ fromAD, rpAD, non-demented controls and Creutzfeldt-JacobDisease (CJD) brains. In addition to commonly knownpartners like Tau and Cellular Prion Protein, we were ableto isolate around 400 different proteins that showed adisease specific involvement. Primary targets were foundto be involved in energy metabolism, neurotransmission,neuronal development and stress response. Assessment ofthe clinical relevance of these aberrations and interactionsis currently underway in various AD models.

Our study presents a novel insight in the Aβ-baseddifferences among slow and rapidly progressive demen-tias, primarily the rapidly progressive variant of AD. Inaddition to providing the evidence that rpAD featuresthe involvement of discrete molecular machinery, italso provides insights into the players involved in pla-cing dementias on a course of rapid progression.

Reference

[1] Portelius, et al. Acta Neuropathol. 2010;120:185–193.

230. Critical structural and kinetic impact of cofactorson host-to-host transmissibility of human prions

Jiri Safar, Chase Kim, Shugui Chen, Tracy Haldiman,Vitautas Smirnovas, Krystyna Surewicz, Nicholar

126 ABSTRACT

Maurer, Qingzhong Kong, Qinzhong Kong and WitoldSurewicz

Case Western, Reserve Univeresity

ABSTRACT

Objectives: Molecular characteristics determiningthe serial host-to-host transmissibility of humanprions causing Creutzfeldt-Jakob disease (CJD) areunknown, and several recent clinical therapeutictrials failed. The key reasons behind these disap-pointing results are (a) fundamental structural differ-ences between human prions causing diseases andcloned laboratory prions, and (b) distinct replicationmechanism of human prions that is not determinedby their confromational stability, as it is in yeast andsome murine prions, but by growth rate of prionaggregates, which is in turn controled by their spe-cific structural features. How these aspects impacttransmissibility of prions to man is an imperativequestion in light of history of iatrogenic and zoonotictransmissions and recent findings of human prions inthe skin and body fluids.Methods: We investigated the effect of distinct cofactorson the structure and replication kinetics of major humanprion strains. Using these data, we synthesized withvariable cofactors a series of new human prions fromthe recombinant human prion protein, and to asses theimpact of glycosylation, monitorred their replicationrate and infectivity in serial passage in transgenic micethat were expressing physiological levels of human prionproteins with or without glycosylation.Results: We report that different lipid and polyanioniccofactors have a major structural and kinetic impact onthe human prion replication in vitro, infectivity in vivo,and strain characteristics. Within the synthetic prionspectrum, recombinant human prion generated withganglioside GM1 cofactor caused unique and stableneurologic dysfunction with incubation times compar-able to natural CJD prion strain in both first andsecond passage, respectively. Unique hybrid MM1/MM2 CJD-like prion neuropathology, high replicationrate of reisolated prions, and biophysical profiling indi-cate that a novel particularly infectious and aggressivehuman prion strain was created.Conclusions: Lipid and polyanion cofactors have funda-mental impact on human prion strain characteristics andtransmisibility. Their seemingly subtle effects on structuralorganization of prions are coupled with large differencesin replication rates, and together with posttranslationalmodifications, play a crucial role as determinants of infec-tivity, host range, and extraordinary precission in target-ting specific brain structures.

231. Prp1 is necessary for seizure-susceptibility inappa zebrafish knockout and new in vivo anti-convulsants

Richard Kanyoa, Laszlo Locskaia, Patricia L. A.Leightona, Harley T. Kuratab and W. Ted Allisona,c

aCentre for Prions & Protein Folding Disease, and Department ofBiological Sciences, University of Alberta, Edmonton, AB, Canada;bDepartment of Pharmacology, University of Alberta, Edmonton,AB, Canada; cDepartment of Medical Genetics, University ofAlberta, Edmonton AB, CANADA

CONTACT Richard Kanyo [email protected]

ABSTRACT

Background: Cellular prion protein (PrPC) has beenlinked to Alzheimer’s disease (AD), the most prevalentdementia. PrPC directly interacts with amyloid-β oligo-mers and amyloid precursor protein (APP) and thegenetic interaction plus mechanism was furtherexplored here. Because 10–22% of AD patients displayepileptic episodes, potential anti-convulsants, ML213and ICA73, which target Kv7.2/3 channels have beeninvestigated in vivo. Zebrafish enable economical high-throughput drug screening platforms that faithfullyrepresent the complexity of the central nervous systemand are therefore an excellent model for these goals.Methods: appa;prp1 compound mutant (Appa andPrp1 are disrupted) fish were engineered by targetedmutagenesis. Seizure-susceptibility was compared inmutant and wild type larvae via treatment with con-vulsants, pentylenetetrazol (PTZ) or 4-aminopyridine(4AP). Locomotor activity was tracked usingEthovision behavioural tracking software. Neural activ-ity of free-swimming animals was measured using thenovel ratiometric fluorescence-based Ca2+ indicatorCaMPARI.Results: PTZ dose-responses of locomotor activityrevealed that Appa and Prp1 single mutants exhibitenhanced seizure-susceptibility compared to wild typefish. However, seizure-susceptibility is dampened/reduced in compound mutants compared to eitherindividual mutant. Measuring neural activity withCaMPARI, and using 4AP as a convulsant, ML213and ICA73 were demonstrated to have anti-convulsantactivity in vivo. ML213 was a very potent anti- convul-sant, whereas ICA73 had similar potency to retigabine(RTG), an FDA approved drug used previously to treatepilepsy. When measuring locomotor activity, ML213and RTG paralleled our results obtained withCaMPARI, but ICA73 did not and displayed anincrease in locomotor activity highlighting the impor-tance of using multiple assays to complement oneanother.

PRION 127

Conclusion: appa and prp1 mutants both exhibitedenhanced seizure-susceptibility. Interestingly, prp1 func-tion was required for maximum seizure-susceptibility inappa mutants suggesting a mechanism/pathway bywhich prp1 functions as a rheostat of seizure-suscept-ibility or neural hyperactivity. Therefore a bi-functionalmechanism may explain our recently published findings[1], where compound prp1;prp2 mutants are more sus-ceptible to seizures than prp1mutants, but less than prp2mutants. ML213 and ICA73 were established here asanti-convulsants in vivo and thus may provide a routein future mechanistic studies to gain sufficient insightand treat AD patients that display seizures. Together,this work using engineered zebrafish and anti-convul-sants could illuminate the PrP-APP pathway as a poten-tial aetiological link between AD and epilepsy.

Reference

[1] Leighton, Kanyo et al. JBC. 2018;293:12576–12592.

232. ‘Novel compounds for the treatment of priondisease’

Robert C.C. Mercera, Nhat T. T. Lea, Doug Fraserb,Aaron Beelerb and David A. Harrisa

aBoston University School of Medicine, Department ofBiochemistry; bBoston University, Department of Chemistry

ABSTRACT

Prion diseases are invariably fatal neurodegenerativediseases of humans and other animals for which thereare no currently available treatments. Using a prioninfected mouse neuroblastoma cell line (ScN2a) as anassay system, many anti-prion compounds have beenidentified, but none have resulted in a clinically effec-tive therapy. Interestingly, while several anti-prioncompounds exert their effects through a direct inter-action with PrPC or PrPSc, the mechanisms underlyingthe anti-prion effects of other compounds remainsenigmatic. A recent report from our laboratory [1]describes the identification of a novel class of anti-prion compounds, phenethyl piperidines. These werediscovered using a high throughput screen for mole-cules that prevent mutant PrP toxicity. JZ107, themost potent derivative, can permanently cure ScN2acells of infection and rescue hippocampal neuronsfrom PrPSc induced synaptotoxicity in vitro. Furtherstudy of JZ107 has led, unexpectedly, to the discoveryof a number of additional, novel anti-prion com-pounds, some of which have previously identifiedmolecular targets and have been used in other clinical

settings. None of these compounds bind directly toPrP, making the identity of their interaction partnersof significant interest. To further validate the anti-prion effects of these compounds, we assessed theirability to prevent the retraction of hippocampal neu-ron dendritic spines following exposure to purifiedprion preparations. We have begun assessing theanti-prion effects of the most potent of these com-pounds in vivo.

Reference

[1] Imberdis T, et al. Identification of anti-prion com-pounds using a novel cellular assay. JBC.2016;291:26164–26176.

233. The many phases of prion protein as observedby NMR

Lauren E. Kleina, Mikail A. Kostelovb, Marcus D.Tuttlea, Stephen M. Strittmatterb and Kurt W. Zilma

aDepartment of Chemistry, Yale University, New Haven,Connecticut, USA; bDepartment of Neurology, Yale School ofMedicine, New Haven, Connecticut, USA

CONTACT Kurt W. Zilm [email protected]

ABSTRACT

Nuclear magnetic resonance (NMR) has proven invalu-able in characterizing all sorts of functionally importantmacromolecular aggregates from amyloids to mem-brane-less organelles. In this contribution we describethe discovery and characterization by NMR of multiplephase states of prion protein [1] with relevance toAlzheimer’s disease (AD). The binding of amyloid-beta oligomers (A β o) to PrPC has been demonstratedto lead to synapse loss and the onset of symptoms in ananimal model of AD. Biophysical investigation of themode of PrPC/A β o association found formation of astoichiometric complex. When studied by magic anglespinning (MAS) NMR, this condensate was revealed tobe a hydrogel phase. We find liquid-like mobility forPrPC in the hydrogel, while the A β o instead behave asa relatively rigid molecular solid. Association of PrPC toA β o is mediated via lysine clusters, which can bedetected by observation of large differential mobilitybetween bound and unbound lysines by 15N MASNMR. Exploration of the PrPC/ A β o phase spacefurther lead to discovery of a PrPC rich viscous liquidphase, also studied by 13C MAS NMR methods.

Repetitive motifs and clustering of identical residuesmakes PrPC a difficult target for NMR resonanceassignment. Nevertheless, this same attribute makes it

128 ABSTRACT

possible to directly associate many resonances withspecific domains. Of the 210 aa in PrPC, 43 are glycines,with all but 3 located in the N-terminal 21–131 seg-ment. Six of the 8 alanines are located in the palindro-mic AGAAAAGA run from 113–120, and 8 of the 12threonines are located at the end of the junctionbetween the C-terminal helices encompassing aa 183–201. Large scale conformational transitions can bedetected by NMR when such aa clusters exhibit simul-taneous shifts characteristic of a change in secondarystructure. In this manner we observe both the N-term-inal fragment and the palindromic sequence to transi-tion from random coil to a largely helical conformationupon A β o binding. Interestingly, the formation of thePrPC liquid phase is found to be accompanied by amelting of the threonine rich helical segment, and theapparent adoption of an unusual χ1 for the tryptophanresidues in the octa-repeat segment.

Biophysical characterization of these phase states ofPrPC is proving relevant to development of possible treat-ments for AD. Compounds that disrupt the hydrogel phasehave been shown to relieve memory deficits in AD mice,and make it possible to detect the selective dissolution ofthe hydrogel phase in Alzheimer’s brain lysates [2].

References

[1] Kostylev MA, et al. Mol Cell. 2018;72(3):426–443.[2] Gunther EC, et al. Cell Rep. 2019;26(1):145–158.

234. The European Union Summary Report OnSurveillance For The Presence Of TransmissibleSpongiform Encephalopathies (TSE): The SituationIn 2017

Angel Ortiz Pelaeza, Valentina Rizzia, Giuseppe Rub,Francesco Ingravalleb and Yves Van der StedeaaUnit on Biological Hazards and Contaminants, Department of RiskAssessment & Scientific Assistance, European Food Safety Authority(EFSA); bBiostatistics, Epidemiology and Risk Analysis (BEAR) Unit.Istituto Zooprofilattico Sperimentale di Piemonte, Liguria e Valle d’Aosta, Torino (Italia)

CONTACT Angel Ortiz Pelaez [email protected]

ABSTRACT

The European Food Safety Authority publishes a yearlysummary of the surveillance activities on transmissiblespongiform encephalopathies (TSE) in bovine animals,sheep, goats, cervids, and other species, in the EuropeanUnion (EU), and in Iceland, Norway and Switzerland.Target groups include: animals clinically suspected ofbeing infected by TSE; animals culled under TSE

eradication measures; animals with clinical signs at ante-mortem; emergency slaughtered; fallen stock/not slaugh-tered for human consumption and healthy slaughteredanimals for human consumption, for cervids also huntedand road or predator-injured or killed.For the first time since bovine spongiform encephalo-pathy (BSE) had been reported, no cases of classicalBSE were reported world-wide in 2017 [1]. Six atypicalBSE cases were reported by Spain (1 H /2 L), France(1 H/1 L) and Ireland (1 L), out of the 1,312,714 cattletested by 28 EU Member States (MS) and 18,526 testedby three non-MS.

In total 431,815 small ruminants were tested in 2017 inthe EU. Comparedwith 2016, there was a 36.2% increase inthe number of cases of classical scrapie (CS) in sheep (933),mostly reported by Greece, Spain, Italy and Romania,although over 75% of the cases were sourced in infectedflocks. Atypical scrapie (AS) was confirmed in 94 animals.In goats, a decrease of 10% in the number of cases ofclassical scrapie (567) were reported, 84% in Cyprus.Atypical scrapie was confirmed in nine animals.

Ten-year trend analysis showed a statistically significantdecrease in the sheep proportion of CS cases per 10,000tested animals and an increase in goats. For AS, 10-yeardata did not detect any statistically significant trend in bothspecies.

After the discovery of chronic wasting disease(CWD) in Norway in 2016, TSE testing in cervidsincreased in the EU: 10 MS tested 3,585 cervids (75%in Romania, 98.5% from wildlife), all negative.Norway tested 25,736 deer in 2017, leading to thedetection of the first case of CWD in a red deer, ninecases in wild reindeer and one in a wild moose.Following EFSA recommendations, the EuropeanCommission introduced a 3-year mandatory surveil-lance programme for six member states starting on 1January 2018.

By the time of submitting this abstract, CS/AS caseswere not yet available, but one new classical BSE casewas confirmed in Scotland (2018), one L-type BSE casein Poland (2019) and one case of CWD in a wild moosein Finland in March 2018.

KEYWORDS: BSE; scrapie; CWD; EU; surveillance

Reference

[1] EFSA (European Food Safety Authority), 2018. TheEuropean Union summary report on surveillance forthe presence of transmissible spongiform encephalopa-thies (TSE) in 2017. EFSA J. 2018;16(11):5492, 64 pp.DOI:10.2903/j.efsa.2018.5492

PRION 129

235. Mutations in the functional amyloid PMELlead to clinical glaucomatous neurodegeneration:Potential for insights into avoiding amyloidosis

W. Ted Allisona,b,c, Tim Footzb, Gavin J Neila,c andMichael A. Walterb

aCentre for Prions & Protein Folding Disease, U Alberta, Edmonton,AB, Canada; bDepartment of Medical Genetics, U Alberta,Edmonton, AB, Canada; cDepartment of Biological Sciences, UAlberta, Edmonton, AB, Canada

CONTACT W. Ted Allison [email protected]

ABSTRACT

Pigmentary glaucoma (PG) is a major form of glau-coma, a leading cause of blindness worldwide.Glaucoma is neurodegeneration of retinal ganglioncells, which are neurons of the CNS. We have, for thefirst time, discovered a causative gene for this CNSneurodegeneration in humans. Our results reveal thatmutations of the premelanosome gene (PMEL) causePG. We identified PMEL mutations in an Albertafamily with PG and in four isolated patients. In parallel,our colleagues Drs. Wiggs (Harvard Medical School)and Craig (U Flinders, Australia), identified PMELmutations in separate PG cohorts, strongly supportingthat PMEL mutations cause PG (Lahola-Chomiaket al. 2018 Human Molecular Genetics doi: 10.1093/hmg/ddy429) [1].

Intriguingly, PMEL protein is a rarity in vertebrates:a functional amyloid. PMEL forms amyloid fibril struc-tures in pigmented cells of vertebrates, includinghumans, and these amyloid fibrils are structurally cri-tical for deposition of melanin pigment. The patientPMEL mutations we identified result in impaired pro-cessing of PMEL protein and altered amyloid structure,however the central question of how these changes leadto CNS neurodegeneration remains unknown. Our zeb-rafish modelling of PMEL loss via CRISPR/Cas9 muta-genesis produced recessive inheritance of swollen eyemorphology in larvae that is strikingly reminiscent ofincreased ocular pressure in glaucoma; this implies thatloss of the functional PMEL amyloid can contribute tosome aspects of PG aetiology. This is in contrast withour patient pedigrees and mutations of PMEL homo-logs in other animals (dogs, horses, etc.) where domi-nant inheritance implies a toxic gain-of-function; thispredicts that abnormal PMEL amyloid might lead toneurodegeneration via a (prion-like?) templating ofaberrant amyloid structure.

Future work is warranted to delineate these contri-butions of gained toxicity vs. reduced protein functionduring (prion-like?) neurodegeneration [2], includingdefining whether both of these contributions are

required for death of CNS neurons. Such work haspotential to give unique insights into amyloidosis: Weidentify several mutations in patients with glaucoma-tous neurodegeneration, and these now guide our pre-dictions of which aspects of PMEL are the key tohuman CNS cells thriving through a lifetime of highamyloid content.

References

[1] Lahola-Chomiak, et al. Hum Mol Genet. 2018.DOI:10.1093/hmg/ddy429

[2] Allison, et al. Int J Mol Sci. 2017;18:E2223.DOI:10.3390/ijms18102223

SCHMITT-ULMS, Gerold 2019

236. A mechanism-based small-molecule approachto the treatment of prion diseases that targets PrPC

MMehrabiana,b, DWilliams a,b, XWanga,b,c, FGhodratia,b,c,M Güneş,a,b and G Schmitt-Ulmsa,b,c

aTanz Centre for Research in Neurodegenerative Diseases,University of Toronto, Toronto, Ontario, Canada; bKrembilDiscovery Centre, Toronto, Ontario, Canada; cDepartment ofLaboratory Medicine & Pathobiology, University of Toronto,Toronto, Ontario, Canada

ABSTRACT

A wide range of observations in humans and animalsindicate that a reduction in steady-state levels of thecellular prion protein (PrPC) is both safe and woulddelay or prevent prion diseases. To derive rationalapproaches for suppressing PrPC levels at the plasmamembrane, we and others have been studying themolecular environment and function of PrPC.

This work led us to recognize that prion genes evolvedfrom a ZIP zinc transporter ancestral molecule with rolesin a morphogenetic programme known as epithelial-to-mesenchymal transition (EMT). The latter guided us toinvestigate the possible involvement of PrPC in EMT,revealing PrPC to control the polysialylation of the neuralcell adhesion molecule 1 (NCAM1), a well-studied post-translational modification that serves as a molecular mar-ker of EMT. In several cell models we interrogated, themolecular environment of PrPC is characterized by itsresidence within a specialized membrane domain thathosts, in addition to NCAM1, distinct subsets of morethan 20 additional proteins. Interestingly, most of theseproteins have known roles in the modulation of TGFB1and integrin sister signalling complexes that are activatedduring EMT.

The binding of ligands to their cognate receptorsoften induces the internalization of ligand-receptor

130 ABSTRACT

complexes, followed by their recycling or degradation.We reasoned that such a turn of events may lead to apassive co-internalization of PrPC, effectively reducingits levels at the cell surface. Our recent data reveal thatPrPC levels are indeed robustly reduced in the presenceof non-toxic nanomolar concentrations of a compound,hereafter referred to as KDC series 100 compound 2(KDC102), which targets one of the proteins in proxi-mity to PrPC. As anticipated, this reduction in PrPC

levels depends on the compound-dependent degrada-tion of the targeted receptor and can be observed in co-cultures of human neurons and astrocytes. Althoughmechanistic insights are not essential for translationinto the clinic, their existence should prove an assetgoing forward. Current efforts are directed towardstesting KDC102 in proof-of-concept prion diseasetreatment studies in rodents.

237. A general mass spectrometry-based methodof quantitating the relative amounts of PrPpolymorphisms in CWD PrPSc

Christopher J. Silvaa, Melissa L. Erickson-Beltranb,Camilo Duque Velásquezc, Judd Aikend and DebbieMcKenziee

aProduce Safety & Microbiology Research Unit USDA, WRRC, ARS;bProduce Safety & Microbiology Research Unit, USDA, WRRC, ARS;cUniversity of Alberta, 120 Brain and Aging Research Building,Edmonton, AB T6G 2R3, Canada; dAgricultural Sciences – Centrefor Prions and Protein Folding Diseases – University of Alberta;eUniveristy of Alberta

ABSTRACT

Background/Introduction: Chronic wasting disease(CWD) was first described in a captive cervid herdin 1967, recognized as a prion disease in 1978, andfirst detected in wild cervids in 1981. CWD is readilytransmitted among animals and from a contaminatedenvironment to uninfected animals. By 2019, 26states in the United States, three Canadian provinces,South Korea, Norway, and Finland had found CWDin captive and/or wild cervid populations. Cervid PrPcontains polymorphisms that influence progression ofCWD and propagation of CWD strains with differenttransmission properties [1–3]. It is important todevelop a method to quantitate the relative amountsof those polymorphisms present in CWD PrPSc, sothat their respective influence on CWD prion propa-gation can be determined.Materials and Methods: Plasmids containing relevantcervid PrP polymorphisms were prepared. The neces-sary 15N-labelled internal standards were produced by

overexpressing the PrP genes in E. coli grown in mini-mal medium with 15NH4Cl as the sole nitrogen source.Peptides derived from the tryptic or chymotrypticdigestion of cervid recombinant PrP and containingthe relevant polymorphisms were identified. A multiplereaction monitoring method (MRM) was developed foreach of those peptides.Results: Calibration curves relating the signal intensityof the various peptides and their absolute amounts asmeasure by protein assay were linear and had excellentcorrelation coefficients. In addition, samples fromexperimentally infected heterozygous deer (Q95S96/Q95G96 and H95G96 /Q95G96) were analysed and therelative amounts were determined. The optimal diges-tion for all tryptic peptides was 20 h. For chymotrypsinthe optimal digestion time was also 20 h, apart from asingle peptide.Conclusions: This approach can be used to quantifythe contribution of specific polymorphisms toPrPCWD formation, enhancing our ability to deter-mine the relative susceptibility of various poly-morphisms to CWD. It can also be used to detectthe distribution of CWD prion strains.

References

[1] Johnson CJ, et al. PLoS One. 2011;6:e17450.[2] Duque Velasquez C, et al. J Virol. 2015;89:12,362–

12,373.[3] Herbst A, et al. Emerg Infect Dis. 2017;23:1598–1600.

238. A general mass spectrometry-based methodof quantitating the oxidation of the methionineside chains in classical and atypical scrapie PrPSc

Christopher J. Silvaa, Melissa L. Erickson-Beltranb,Inmaculada Martin-Burrielc, Juan José Badiolad, JesusR. Requenae and Rosa Boleaa,f

aProduce Safety & Microbiology Research Unit, USDA, WRRC, ARS;bProduce Safety & Microbiology Research Unity, USDA, WRRC, ARS;cUniversity of Zaragoza, Zaragoza, Spain; dUniversity of Zaragoza;eUniversity of Santiago de Compostela; fUniversity of Zaragoza –Centre for TSE and emerging animal diseases

ABSTRACT

Background/Introduction: The structure of PrPSc isuncertain, but the most compelling data suggests thatit is a four-rung β -solenoid. The rungs of the solenoidare β -sheet secondary structures held together byhydrogen bonds. Unstructured loops connect themore rigid β -sheet structures. The side chains of theamino acids in the β -sheets alternately project inside

PRION 131

the solenoid or outside of it. This may make the reac-tivity of adjacent amino side chains dramatically differ-ent. The side chain of methionine can be oxidizedunder physiological conditions to the much morepolar sulfoxide. Eukaryotic cells possess enzymes thatreduce oxidized methionines, so the methionines ofPrPC should be largely unoxidized. Methionines pre-sent in PrPSc may be oxidized by chemical reagents ornatural oxidative processes. Quantitating the extent ofthis oxidation can provide insight into the location ofthe methionines within the β -solenoid.Materials and Methods: Plasmids containing relevantPrP sequences were prepared.1 15N-labelled internalstandards were prepared by overexpressing the PrPgenes in E. coli grown in minimal medium with15NH4Cl as the sole nitrogen source. A set of peptidescontaining the methionines was identified that arederived from the tryptic or chymotryptic digestion ofsheep PrP. A multiple reaction monitoring method(MRM) was developed to detect and quantitate eachpeptide.Results: Calibration curves relating signal intensities ofthe various peptides and their absolute amounts werelinear and had excellent correlation coefficients. Theoptimal digestion for all tryptic peptides was 20 h. Forchymotrypsin the optimal digestion was 20 h, exceptfor one peptide. In peptides containing two methio-nines, the oxidation state of each could be readilyidentified by a combination of chromatographic prop-erties and the MRM method.Conclusions: This approach can be used to quantitatethe relative amounts of each oxidized methionine insheep scrapie. By comparing the results obtained fromclassical and atypical scrapie, we gained insight into thestructural differences between the two isoforms.

239. Hsp110 modifies prion infection in vitro andin vivo

Cristóbal Marrero-Winkensa,b,c and Hermann M.Schatzla,b,c

aDepartment of Comparative Biology and Experimental Medicine,Faculty of Veterinary Medicine, AB, Canada; bCalgary Prion ResearchUnit, AB, Canada; cHotchkiss Brain Institute, University of Calgary,AB, Canada

CONTACT Cristóbal Marrero-Winkens [email protected]

KEYWORDS: Hsp110; chaperones; prion replication;prion fragmentation; disaggregase function

Mammalian heat shock protein 110 (Hsp110) familymembers cooperate with Hsp70 and Hsp40 to disag-gregate misfolded proteins. Efficient in vitro

disaggregation of α-synuclein and artificially denaturedproteins by this tri-chaperone system has recently beenreported. In yeast, it is well established that the disag-gregase formed by Hsp70 and Hsp104 (which has nomammalian homolog) affects the maintenance of prionphenotypes. We hypothesize that the mammalian tri-chaperone system fulfils a similar function by partici-pating in prion fragmentation and disaggregation. Totest this, Hsp110 levels were transiently manipulated in22L-ScN2a cells. Transient knockdown reduced thelevels of PrPSc, while overexpression led to a dose-dependent increase in PrPSc. These effects are reminis-cent of Hsp104 modulation in yeast. Next, Hsp110overexpressing and wild-type mice were inoculatedwith 22L and Me7 prions. Overexpression of Hsp110significantly prolonged the survival of Me7- but not22L-inoculated animals. Sucrose gradient ultracentrifu-gation of brain homogenates derived from terminallydiseased animals also revealed a trend towards largerPrPSc aggregates in overexpressing mice. Overall, theseresults confirm that Hsp110 is involved in PrPSc frag-mentation and warrant further study. Finally, we alsoreport on on-going studies, which include prion inocu-lation of different overexpressing mice as well as thegeneration of Hsp110 knockout and overexpressing celllines using the CRISPR-Cas9 system and recombinantlentiviruses

240. The NLRP3-Caspase 1 Negatively RegulatesAutophagy via TLR4-TRIF in Prion Peptide-InfectedMicroglia

Mengyu Lai†, Hao Yao†, Syed Zahid Ali Shah, DemingZhao and Lifeng Yang

National Animal Transmissible Spongiform EncephalopathyLaboratory, College of Veterinary Medicine, China AgriculturalUniversity, Beijing, China

CONTACT Lifeng Yang [email protected]

†Deceased.

ABSTRACT

Introduction: Prion diseases are neurodegenerative disor-ders characterized by the accumulation of misfolded prionprotein, spongiform changes in the brain, and braininflammation as a result of the wide-spread activation ofmicroglia. Autophagy is a highly conserved catabolic pro-cess for the clearance of cytoplasmic components, includ-ing protein aggregates and damaged organelles, thisprocess also eliminates pathological PrPSc as it accumulatesduring prion infection [1]. The NALP3 inflammasome is amultiprotein complex that is a component of the innateimmune system and is responsible for the release of pro-

132 ABSTRACT

inflammatory cytokines. Our previous study showed thatthe neurotoxic prion peptide PrP106-126 induces NALP3inflammasome activation and subsequent IL-1β release inmicroglia [2]. Autophagy is involved in the regulation ofthe immune responses and inflammation in many diseasesincluding neurodegenerative diseases. However, the rela-tionship between autophagy and NALP3 inflammasome inprion diseases has not been investigated.Results: In this study, we demonstrated that the proces-sing and release of mature IL-1β is significantly enhancedby the inhibition of autophagy. Conversely, gene silencingof the NALP3 inflammasome promotes autophagy.Suppression of TRIF or TLR4 by siRNA attenuatedPrP106-126-induced autophagy, which is indicating thatthe TLR4-TRIF signalling pathway is involved in PrP106-26-induced autophagy. Caspase 1 directly cleaved TRIF todiminish TLR-4-TRIF mediated autophagy. Our findingssuggest that the inhibition of autophagy by NALP3inflammasome is probably mediated by activatedCaspase-1-induced TRIF cleavage [3]. This is the firststudy reporting that the NALP3 inflammasome complexnegatively regulates autophagy in response to PrP106-126stimulation in microglia, and partly explains the mechan-ism of autophagy inhibition by Caspase-1 in PrP106-126-induced BV2 cell activation.Conclusions: Our findings suggest that autophagy up-regulation and inhibition of Caspase-1 may protectagainst prion-induced neuroinflammation and acceler-ate misfolded protein degradation and are potentialtherapeutic approaches for prion diseases.

References

[1] Deretic V, et al. Autophagy in infection,inflammationand immunity. Nat Rev Immunol 2013;13:722–737.

[2] Shi F, et al. Inhibition of phagocytosis and lysosomalacidification suppresses neurotoxic prion peptideinduced NALP3 inflammasome activation in BV2microglia. J Neuroimmunol. 2013;260:121–125.

[3] Faure M., et al. Pathogen-induced autophagy signalingin innate immunity. J Innate Immun. 2013;5:456–470.

241. Evaluating the applicability of tauK18 as RT-QuIC reaction substrate for the analysis of samplescollected from patients with neurodegenerativedisorders

Martina Rossia, Chiara Maria Giulia De Lucaa, Elena DeCeccoa, Joanna Narkiewicza, Giulia Salzanoa, SaraMaria Portaleoneb, Antonio Emanuele Eliac, RobertoEleoprac, Giuseppe Legnamea, Giorgio Giacconed andFabio Modad

aLaboratory of Prion Biology, Department of Neuroscience, ScuolaInternazionale Superiore di Studi Avanzati, Trieste, Italy;bNeuropathology-Neurology 5 Unit, Fondazione IRCCS IstitutoNeurologico Carlo Besta, Milan, Italy; cMovement DisordersDepartment – Neurology I Unit, Fondazione IRCCS IstitutoNeurologico Carlo Besta, Milano, Italy; dOtolaryngology Unit, SanPaolo Hospital, Department of Health Sciences, University of Milan,Milan, Italy

CONTACT Martina Rossi [email protected]

ABSTRACT

Introduction: Neurodegenerative diseases are charac-terized by the accumulation of abnormally folded pro-teins in different regions of the brain [1,2]. Amongthem, Alzheimer’s disease (AD), Frontotemporaldementia (FTLD-tau), Corticobasal Degeneration(CBD), and Progressive Supranuclear Palsy (PSP) areassociated to different abnormal conformations of themicrotubule-associated tau protein. Clinical diagnosisof tauopathies is often challenging due to their over-lapping symptoms, especially in the early stages [2].Compelling evidence suggest that abnormally foldedproteins appear in peripheral tissues of dementedpatients [3–5]. Therefore, we are currently exploitingthe Real-Time Quaking Induced Conversion (RT-QuIC) assay to attempt detection of abnormal tau inperipheral tissues of patients with tauopathies.Materials and methods: Recombinant tau fragmentcomprising the 4-repeats domain (tauK18) was usedas substrate for RT-QuIC assay. In vitro producedtauK18 aggregates (referred to as artificial seeds) wereserially diluted and added to new RT-QuIC reactions toassess its sensitivity. Similarly, brain homogenates(BHs) collected from patients with PSP, FTLD-tauand AD were analysed by means of RT-QuIC. BHscollected from patients with other dementias (includingParkinson’s disease, Dementia with Lewy bodies andMultiple System Atrophy) and subject not affected byneurodegenerative disorders (NDP) were used as con-trols. Olfactory mucosa (OM) samples collected frompatients with a clinical diagnosis of CBD, PSP, and ADare currently under RT-QuIC analysis. OM from PD,MSA, DLB patients, and healthy subjects are includedas controls.Results: All dilutions of artificial seed efficiently accel-erated the kinetics of tauK18 aggregation, thus reveal-ing the ability of RT-QuIC in detecting up to attogramsof tauK18 aggregates. BHs of patients with PSP, FTLD-tau and AD accelerated tauK18 aggregation withgreater efficiency than that of DLB, PD, MSA, andNDP. Preliminary data collected from RT-QuIC analy-sis of OM samples suggest that the aggregation of

PRION 133

tauK18 is more efficient when the reaction is supple-mented with CBD and PSP samples. Less efficientaggregation was observed after the addition of theother OM samples.Conclusions: The aggregation kinetics of tauK18 used asRT-QuIC reaction substrate can be influenced by otherfactors other than tau itself, therefore representing animportant issue especially from a diagnostic point ofview. The presence of other amyloidogenic proteins (e.g.α-synuclein or β-amyloid) can play a pivotal role in pro-moting tauK18 aggregation, thus generating misleadingresults. For this reason, we are currently coupling RT-QuIC analysis with structural and biochemical studies toassess whether the final reaction products acquire peculiarfeatures useful for disease discrimination.

KEYWORDS: tauK18; RT-QuIC; diagnosis;tauopathies

References

[1] Soto C. Nat Rev Neurosci. 2003;4:49.[2] Irwin DJ. Parkinsonism Relat Disord. 2015;22 Suppl 1

(01):S29–S33.[3] Moda F. et al., N Engl J Med. 7 August 2014;371

(6):530–539.[4] Saijo E. et al., Acta Neuropathol. 2017 May;133(5):751–

765.[5] Redaelli V, Bistaffa E. et al., Sci Rep. 7 April

2017;7:46269.

242. Incubation periods of classical and atypicalbovine spongiform encephalopathies (BSE) reflectan inverse relationship between PrPSc-associatedneuroinflammation and autophagy

NajibaMammadovaa,b,c, M. HeatherWest Greenleeb,c,d, S.Jo Mooree, Donald S. Sakaguchia,c and Justin J. Greenleee

aDepartment of Genetics, Development and Cell Biology, IowaState University, Ames, IA, USA; bImmunobiology GraduateProgram, Iowa State University, Ames, IA, USA; cNeuroscienceGraduate Program, Iowa State University, Ames, IA, USA;dDepartment of Biomedical Sciences, Iowa State UniversityCollege of Veterinary Medicine, Ames, IA, USA; eVirus and PrionResearch Unit, National Animal Disease Center, USDA, AgriculturalResearch Service, Ames, IA,USA

CONTACT M. Heather West Greenlee [email protected]

ABSTRACT

Neurodegenerative protein misfolding disorders are agroup of diseases in both humans and animals with asimilar mechanism of disease progression. These dis-eases, that include Alzheimer’s disease, Parkinson’sdisease, chronic wasting disease, and bovine

spongiform encephalopathy (BSE) result from aber-rant folding and accumulation of disease specificproteins. Transmissible spongiform encephalopathies(TSEs) are diseases that result from improper foldingand accumulation of the prion protein. Due to theirtransmissibility, TSEs are an important experimentalmodel to study the basic pathobiology of all proteinmisfolding disorders. TSE strain variations can influ-ence disease phenotypes such as host susceptibility,biochemical and immunohistochemical profiles, andincubation periods. BSE is a TSE that occurs in cattlethat can be subdivided into three different strains:classical BSE, atypical high (H)-type, and atypical low(L)-type BSE. Both H-type and L-type BSEs, haveshorter incubation periods after experimental inocu-lation and, therefore, an accelerated disease progres-sion when compared to classical BSE. Currently,there is a lack of knowledge about the factors thatinfluence accumulation of misfolded proteins anddisease progression making this a key challenge forthe development of therapies for protein misfoldingdiseases. In this study, we used the differencesbetween classical (n = 12) and atypical BSEs(n = 20) as a model to identify the molecular factorsassociated with disease progression. The NLRP3inflammasome is a critical component of the innateimmune system that leads to release of IL-1β(Interlukin-1β), an important regulator of neuroin-flammation in many protein misfolding diseases.Macroautophagy is an intracellular mechanism thatplays an essential role in protein clearance andhomeostasis. Here we used immunohistochemistryto assess the retina and three brain regions (thala-mus, caudate nucleus, and brainstem at the level ofthe obex) to investigate the relationship between dis-ease incubation period, PrPSc accumulation, neuroin-flammation, and changes in macroautophagy. Wedemonstrate that atypical BSEs, characterized byshorter incubation periods, present with greater accu-mulation of PrPSc, glial-cell activation, NLRP3inflammasome activation, and decreased autophagy.Our work demonstrates a relationship between dis-ease time course, neuroinflammation, and the autop-hagic stress response, that has not been previouslyreported. This work may help identify novel thera-peutic approaches that can delay or even prevent theprogression of protein-misfolding diseases.

243. Comparative effect of green tea polyphenolson the amyloid aggregation of human prionprotein

Nikita Admane and Abhinav Grover

134 ABSTRACT

School of Biotechnology, Jawaharlal Nehru University, NewDelhi, USA

CONTACT Nikita Admane [email protected]

ABSTRACT

Misfolding and aggregation of human prion protein(HuPrP) is a hallmark to a variety of neurologicaldisorders, commonly known as the TransmissibleSpongiform Encephalopathies (TSEs). Despite severalefforts to recognize effective small molecule inhibitorsof amyloid disorders, presently no effective therapeuticagent is available for the neurodengerative amyloidosis.Herein, we examined the protective efficacy of twogreen tea polyphenols, GTP1 and GTP2 against theamyloid conversion of cellular human prion proteinto the disease forming scrapie conformation, employinga combination of computational, and experimentalapproach. Collective results from ThT, TEM, AFM,DLS, and CD experiments show that both the GTPsexhibit an inhibitory effect on the structural transitionof HuPrP monomers into beta sheet rich structures,thereby reducing the amyloid fibrillization of HuPrP.Further, MTT assay and confocal microscopy revealedthat both the catechins increase the cell viability byreducing toxicity associated with oligomeric species inneuronal cell models. But interestingly, GTP1 has astronger inhibitory effect on HuPrP amyloidogenesisas compared to GTP2 due to minor differences in thestructure of these two polyphenols. Both the com-pounds stably interact with the hydrophobic residuesin the binding pocket of HuPrP which encompasses theβ2-α2 loop and the C-terminal region of the α3 helix.Binding mechanisms of GTP1 and GTP2 with HuPrPwas analysed by molecular dynamics simulations. The4Å region of HuPrP binding pocket around these twocompounds reveals that GTP1 forms more hydrogenbonds and hydrophobic interactions (as compared toGTP2) with the residues critical in the conversion ofcellular to scrapie form of HuPrP, this may be thereason behind making GTP1 a more potent inhibitor.Thus, small molecules (like catechins and polyphenols)that can directly interact with the critical hydrophobicpatches of HuPrP can act as inhibitors of prion amy-loidogenesis and hold a substantial therapeutic poten-tial. The green tea polyphenols might also be ofsynergistic benefit in treating the prion diseases.

244. The use of recreational hunters in ChronicWasting Disease (CWD) management: behaviourand incentives

Lusi Xiea, Vic Adamowicza and Patrick Lloyd-Smithb

aUniversity of Alberta; bUniversity of Saskatchewan

CONTACT Lusi Xie [email protected]

ABSTRACT

Background:While many jurisdictions in NorthAmerica have implemented some management pro-grammes to address Chronic Wasting Disease (CWD),the disease continues to spread. Although epidemiolo-gical models have proposed depopulating infected ani-mals to reduce disease prevalence and spread byincreasing hunter harvest (e.g. Potapov et al. 2016)[1], these models do not evaluate the incentivesrequired to increase hunter harvest or the impacts ofwildlife disease and management programmes onrecreational activity and value. A better understandingof human behavioural responses will help effectivelyincorporate recreational hunters in wildlife disease con-trol. The objective of this study is to examine the use ofincentives to recreational hunters for CWD manage-ment by studying hunting trip decisions in response toCWD and its management. We develop an empiricalmodel, with a focus on spatial and temporal substitu-tion patterns of recreational activities, as well as eco-nomic benefits of a new recreation season.Materials and Methods:The data used in this study arefrom an online survey of recreational hunters inAlberta and Canada. Five thousand hunters weresampled and a response rate of approximately 20%resulted in a usable sample of 707. Hunters wereasked to recall hunting trips in 2017 and statedintended visitation with proposed extended huntingseasons for CWD management. We develop a dis-crete-continuous empirical model that captures hunt-ing trip decisions on sites, periods and frequencies.Explanatory variables included in the model are CWDprevalence levels, policy dummy variables, individuals’socio-demographic variables as well as travel and timecosts. With the estimated parameters, spatial and tem-poral substitution patterns are captured, measures ofthe potential harvest of affected deer populations, andeconomic benefits, are calculated.Results and Conclusions:We find a negative but insig-nificant coefficient of CWD which indicates that indi-viduals do not avoid hunting in CWD-infected areas.Individuals are likely to take around six additional tripson average during the extended seasons – this is morethan half of the average trips (around 10) they actuallytook in 2017. We find that individuals are likely tosubstitute from hunting in areas with lower CWDrisks to hunting in the most CWD-infected areas inboth seasons. Individuals are better off from anextended season for CWD management. Our findings

PRION 135

indicates that it is possible to direct hunting trips forCWD control by extending hunting seasons in CWD-infected areas without additional monetary or non-monetary incentives.

KEYWORDS: Chronic wasting disease; recreationdemand

Reference

[1] Potapov, et al. PLoS One. 2016;11(3):e0151039.

245. PrPC knockdown by Liposome-siRNA-peptidecomplexes (LSPCs) prolongs survival and normalbehaviour in prion-infected mice immunotolerantto treatment

Heather Bendera,b, Noelle Noyesa,c, Jessica L Annisa,Amanda Hitpasa, Luke Mollnowa, Kendra Croaka,Sarah Kanea, Kaitlyn Wagnera, Steven Dowa,d andMark D. Zabela

aPrion Research Center, Department of Microbiology, Immunologyand Pathology, College of Veterinary Medicine and BiomedicalSciences, Colorado State University, Fort Collins, CO, USA;bDepartment of Pharmacology, University of Colorado School ofMedicine, Aurora, CO, USA; cDepartment of Veterinary PopulationMedicine, College of Veterinary Medicine, University of Minnesota,St. Paul, MN, USA; dCenter for Immune and Regenerative Medicine,Department of Clinical Sciences, Colorado State University, FortCollins, CO, USA

CONTACT Mark D. Zabel [email protected]

ABSTRACT

Prion diseases are members of neurodegenerative, pro-tein misfolding diseases (NPMDs) that includeAlzheimer’s, Parkinson’s and Huntington diseases,Amyotrophic Lateral Sclerosis, tauopathies, traumaticbrain injuries, and chronic traumatic encephalopathies.No known therapeutics improve quality of life or sur-vival times of humans afflicted with prion disease. Weand others developed a new approach to NPMD ther-apy based on reducing the amount of the normal, host-encoded protein that acts as a substrate for misfoldinginto pathologic forms using RNA interference, a path-way that decreases levels of mRNA encoding a parti-cular protein. We developed a non-invasive therapeuticdelivery system consisting of siRNA complexed to lipo-somes and addressed to the central nervous systemusing a targeting peptide derived from Rabies virusglycoprotein and delivered transvascularly. These lipo-some-siRNA-peptide complexes (LSPCs) cross theblood-brain barrier and deliver PrP siRNA to neuronalcells to decrease expression of the normal cellular prion

protein, PrPC, which acts as a substrate for prionreplication. Here we show that LSPCs can extend sur-vival and improve behaviour of prion-infected micethat remain immunotolerant to treatment. LSPC treat-ment may be a viable therapy for prion and otherNPMDs that can improve the quality of life of patientsat terminal disease stages.

246. Congenital hydrocephalus with drainageproblems leading to CJD and Abeta accumulation.A case report

Annemieke Rozemuller, Wim Spliet, Anja Horstman,Will Hermsen, Vandana Gupta and Wim van Hecke

Prionlab. Dept. of Pathology, University Medical Center, Utrecht,The Netherlands

CONTACT Annemieke Rozemuller [email protected]

Creutzfeldt-Jakob *amyloid beta *hydrocephalus*drainage.

ABSTRACT

Introduction: We describe the neuropathology of a 48-year-old man with congenital hydrocephalus and CSFdrain from his 18th year who became slow and gotrapidly progressive dementia in 2 months, with epi-lepsy, dysarthria, gait abnormalities and myoclonus.The EEG was typical for CJD. The RT-QuIC test waspositive.M&M: The brain autopsy was performed in the prion-lab UMCUtrecht. The brain weight was 1410 g; thedrain was situated in the bottom of the fourth ventricle.No hydrocephalus was seen but the aquaduct was extre-mely small. Twenty-five blocks were taken from rele-vant areas and stained for HE, Kluver-PAS as wellimmunohistochemically for 3F4. Next to that westained for Abeta and tau.Results: Routine staining showed fine vacuolar spon-giosis of the grey matter and prion staining fitted with ahistotype of MM/MV1 according to the histotyping ofParchi et al. (2012). In addition numerous diffuse Abetaplaques were found in the cortex but no classical amy-loid plaques or (pre)tangles in the tau staining. Just afew dystrophic neurites.Conclusion: This case report shows that congenitalhydrocephalus with drainage problems can lead to pro-tein misfolding with both prion accumulation(Creutzfeldt – Jakob disease) and numerous Abeta pla-ques on the age of 48 year. This supports earlier the-ories that drainage is essential for removing misfoldedproteins.

136 ABSTRACT

247. Use of modern population genetic techniquesto investigate the effects of the epidemic of kuru, aunique prion disease in the Papua New Guineahighlands

Liam Quinna, Jerome Whitfielda, Garrett Hellenthalb,Michael Alpersc,d, John Collingea, and Simon Meada

aMRC Prion Unit at UCL, UCL Institute of Prion Diseases, London,UK; bDepartment Genetics, Evolution & Environment, UniversityCollege London; cCentre for International Health, CurtinUniversity, Perth, Australia; dPapua New Guinea Institute ofMedical Research, Goroka, Papua New Guinea

CONTACT Liam Quinn [email protected]; Jerome [email protected]

ABSTRACT

Background: Kuru was the first observed and to thisday remains the largest human prion disease epidemic.It devastatingly affected the Fore linguistic and nearbygroups in the Papua New Guinea (PNG) highlands. Thecondition was transmitted by consumption of prioninfected material at mortuary rituals. Over the first 20documented years of the epidemic ~2,400 people diedfrom kuru in communities with populations of ~40,000individuals. The Eastern Highlands of Papua NewGuinea where the Fore speaking communities resideis marked for its cultural and linguistic diversity.During the kuru epidemic the region was undergoinga marked transition with European contact and colonialadministration. These factors present challenges tofurther understanding the population response tokuru specifically and approaches to find novel variantsthat affected disease predisposition.Materials and Methods: We analysed a unique genomewide array dataset of 1,513 individuals from the affectedcommunities from 22 linguistic groups and 50 across theEastern Highlands of PNG. We used population genetictechniques including Chromopainter analysis andfineStructure clustering to understand the populationstructure of the region, analyse patterns of gene flowand migration, tested for evidence of historical admixtureevents and evidence of genetic signatures of the impact ofkuru on the Fore region including the marked sex bias incases observed during the epidemic.Results: We find a strong population structure withPrincipal Component Analysis and fineStructure clusteringshowing a strong tendency for individuals to cluster withindividuals from the same linguistic grouping. Mean FSTvalues observed in the Eastern Highlands were 0.02, fargreater than those observed in the United Kingdom(0.0007) reflecting the degree of distinction betweengroups. Although we find that linguistic group member-ship explains many aspects of population structure we also

find that topographical features and cultural differencesalso exert strong forces. Gene flow between linguisticgroups is evident with the presence of individuals reflectinghistorical processes of short and long distance migration.Conclusion: We show that the affected populationshave displayed remarkable resilience with no percepti-ble derivation in population genetic parameters overalldespite facing such an existential threat. The markedpopulation structure in the region and processes ofgene flow present challenges for ongoing investigationsinto the genetic response and genetic architecture ofkuru disease.

KEYWORDS: Kuru; population genetics; natural selec-tion; genetic epidemiology

248. How the PrPC C-Terminal, globular domainregulates its toxic N-Terminal Domain

Glenn L. Millhausera, David A. Harrisb, GrahamRosemana, Kevin Schillinga, Kate Markhama, AlexMcDonaldb and Bei Wub

aDepartment of Chemistry & Biochemistry, UC Santa Cruz, SantaCruz, CA, USA; bDepartment of Biochemistry, Boston UniversitySchool of Medicine, Boston, MA, USA

CONTACT Glenn L. Millhauser [email protected]

ABSTRACT

The cellular prion protein (PrPC) is composed of twoprimary segments – a helical C-terminal domain and aflexible N-terminal domain. Research over the last5 years finds that the N-terminal domain is responsiblefor the monomeric protein’s neuronal toxicity asrevealed by toxic, spontaneous, electrophysiologicalcurrents and antibody induced neuronal death in cul-tured cells and brain slices. This inherent toxicity isarrested, however, by the regulatory C-terminaldomain. Our research is focused on revealing the mole-cular mechanism underlying this critical regulatoryfunction. The physiologic metal ions copper and zincboth bind to the octapeptide repeat segments in the N-terminal domain. Using advanced magnetic resonanceapproaches, along with electrophysiology measure-ments, we have identified a previously unseen intramo-lecular switch in PrPC whereby copper or zinc drive atertiary contact between the N-terminal domain and anegatively charged, highly conserved surface on the C-terminal domain. Consistent with this model, we findthat amino acid alterations at the docking surface thatdisrupt metal ion binding lead to toxic transmembranecurrents. Together, our research provides a new para-digm for understanding PrPC toxicity and how familialmutations cause prion disease.

PRION 137

Funding

Supported by NIH grants R01 GM06790 (GM), S10OD018455 (GM) and R01 NS065244 (DH).

References

[1] Spevacek, et al. Structure. 2013;21:236–246.[2] Evans, et al. Structure. 2016;24:1057–1067.[3] Wu, et al. eLife. 2017. DOI:10.7554/eLife.23473.001

249. Validation studies of bovine heparinmanufacturing process spiked with BSE agent

Cyrus Bett, Omozusi Andrews, Teresa Pilant, David M.Asher and Luisa Gregori

FDA/CBER/OBRR/DETTD/LBTSEA, Silver Spring, MD, USA

CONTACT Cyrus Bett [email protected]

ABSTRACT

Heparin is an injectable anticoagulant marketed in theUS since the 1930’s and used by 12 million Americansevery year. Heparin is usually processed from mucosaof porcine or bovine intestines. In 2000, US manufac-turers, concerned about possible contamination ofbovine tissues with bovine spongiform encephalopathy(BSE) agent, stopped manufacturing bovine heparin.Thus, only porcine heparin is now marketed in theUS, and about 75% originates from a single foreigncountry. Alternative sources of crude heparin wouldalleviate ongoing concerns about relying on singlecountry and single animal species as a source of allheparin. This would also mitigate the risk of potentialfuture shortages and episodes of adulteration, observedin the past. Bovine heparin is an obvious option, butBSE agent clearance by the bovine heparin manufactur-ing process should be evaluated. To this end, weapplied a four-step bench-scale heparin purificationprotocol resembling a typical heparin manufacturingprocess to investigate removal of the spiked BSEagent. We removed aliquots from each step and ana-lysed them for residual abnormal prion protein(PrPTSE) using real-time quaking-induced conversion(RT-QuIC) assay, and for infectivity using animalbioassays. The purification process reduced infectivityand PrPTSE by more than 3 logs. These findings showthat the heparin manufacturing process has the capacityto remove substantial amounts of the BSE agent andsupport efforts to reintroduce safe bovine heparin inthe USA.

250. Interaction with phosphatidic acid leads toprion protein fibrilization

R. F. D. Santosa,c, C. Alvesa,b, L. V. Menezesb,c, J. M. A.Britoa,c, J. L. Silvaa,c and T. C. R. G. Vieiraa,c

aMedical Biochemistry Institute Leopoldo De Meis, FederalUniversity of Rio de Janeiro; bFederal Institute of Rio de Janeiro;cNational Institute of Cience and Tecnology of Estructural Biologyand Bioimaging, Brazil

ABSTRACT

Transmissible Spongiform Encephalopathies (TSEs)are neurodegenerative diseases caused by misfoldingof the prion protein (PrP), which assumes a patho-genic conformation and propagates the infectionthrough the conversion of native proteins. Recentmodels indicate the possible role of biological ligandsin this process, which would act as cofactors in theconversion reaction. Lipids present in the mamma-lian cell membrane, especially in brain cells, havebeen suggested as endogenous adjuvants promotingprion aggregation. Despite this, little is known aboutthe physical and chemical characteristics of this inter-action. This study investigates the interaction andstructural changes of recombinant PrP (murine, ham-ster and rabbit) in the presence of Phosphatidic Acid(PA) lipid vesicles, in an in vitro model. Full lengthor 90–231 fragment of rPrPC were incubated with PAvesicles. The samples were then analysed by spectro-scopic techniques to detect changes in aggregationand tertiary structure. Increasing concentrations ofPA raised rPrP light scattering, followed by changesin tryptophan fluorescence, indicating the formationof large aggregates. Interaction with PA induced anincrease in beta-sheet content, with an intermediateenrichment compared to PrPSc. These aggregates hadan amorphous and amyloid fibre structure, shown bythioflavin T binding and microscopy observation. PrPtruncated form showed more tendencies to formfibrils. rabPrP:PA aggregation was less robust thatthe other constructs and pH dependent. The resultsindicate that PA vesicles interact with rPrP, leadingto the formation of amorphous aggregates and amy-loid fibrils, close to PrPSc secondary structure. Theabsence of the N-terminal is important to follow afibrillization pathway induced by PA. rabPrP is ableto interact with the biological cofactor tested, butwith less effect depending on pH, suggesting thatthe different binding sites exposed at these pHs canbe important.

KEYWORDS: Prion; neurodegenerative diseases;lipids; aggregation; vesicles

138 ABSTRACT

Sponsorship: FAPERJ, CNPq, CAPES, IFRJ, INBEB

251. The photodynamic inactivation reduces prioninfectivity in phthalocyanine treated RML brainhomogenate

Marie Kostelanskaa, Zdenka Backovska Hanusovaa,Jakub Soukupa, Radoslav Matejb, Karel Holadaa

aInstitute of Immunology and Microbiology, First Faculty ofMedicine, Charles University and General University Hospital inPrague, Czech Republic; bDepartment of Pathology and MolecularMedicine, Thomayer Teaching Hospital, Prague, Czech Republic

CONTACT Marie Kostelanska [email protected]

ABSTRACT

Introduction: Prions are highly resistant to conven-tional sterilization procedures and currently recom-mended inactivation methods include treatment withhighly corrosive chemicals like 2% NaClO or 1MNaOH. Such harsh treatment is incompatible with anumber of delicate medical tools. We have previouslydemonstrated that nontoxic disulfonated hydroxyalu-minum phthalocyanine (AlPcOH(SO3)2) can be usedto initiate photodynamic inactivation (PDI) of prionsat ambient pressure and temperature.1 The aim of thisstudy was to demonstrate the effectiveness of PDI ofprions by bioassay in laboratory mouse.Methods: The PDI was performed on 1% RML oruninfectious CD1 brain homogenates (BH) treatedwith 25 µg ml−1 of AlPcOH(SO3)2 and exposed to redlight (18 min, 4 ×2.5W LED). Control RML inoculumwas prepared similarly but was kept in the dark. Inmouse bioassay, CD1 female mice (n = 10) were intra-cerebrally inoculated with a 25-µl aliquot of AlPcOH(SO3)2-light treated RML BH or control RML BH con-taining AlPcOH(SO3)2 but not irradiated by light.Other controls included mice (n = 5) inoculated withuntreated, uninfectious CD1 BH or CD1 homogenatecontaining AlPcOH(SO3)2, either irradiated or nonirra-diated. For comparison groups of mice (n = 5) wereinoculated with 10-fold dilutions of RML BH (10−2 to10−6). The animals were sacrificed after reaching theterminal disease phase. The presence of PrPres inmouse brains was detected by WB and evaluation ofPrPres distribution in brains by immunohistochemistryis ongoing.Results: Mice inoculated with PDI-treated RML BH (ata 10−2 dilution) survived significantly longer (204 ±23 days) than mice inoculated with the homogenatecontaining an identical amount of AlPcOH(SO3)2,that was not exposed to light (165 ± 11 days). Theirsurvival was also significantly longer than that of mice

inoculated with straight 10−2, 10−3, and 10−4 dilutionsof the RML BH, who survived 169 ± 11, 165 ± 12 and176 ±15 days, respectively. The transmission rate was100% in all groups of mice including serial dilution(10−2 to 10−6). A comparison of survival time-basedon the regression line suggested a decrease in theprion infectivity titer of the RML BH of more than 4orders of magnitude after PDI treatment.Conclusions: Our results suggest that PDI withAlPcOH(SO3)2 leads to substantial decrease (~4 log10)of prion infectivity in RML BH. The PDI of prions bylight induced photocatalytic activity of phthalocyaninesrepresents promising way of prion decontamination.

KEYWORDS: Prion; phthalocyanine; photodynamicinactivation; decontamination; mouse bioassay

Funding

The project was supported by AZV NV18-04–00179.

Reference

[1] Janouskova, et al. J Gen Virol. 2012;93(Pt11): 2512–7.

252. RT-QuIC as an antemortem diagnostic tool todetect chronic wasting disease in deer skin

Natália C. Ferreiraa, Jorge M. Charcob, Michael A.Metricka, Christina D. Orrua, Andrew G. Hughsona,Joaquín Castillaa, Michael W. Millerc and ByronCaugheya

aLaboratory of Persistent Viral Diseases, Rocky MountainLaboratories, National Institute of Allergy and Infectious Diseases,National Institutes of Health, Hamilton, MT, USA; 2 CIC bioGUNE,Derio (Bizkaia), Spain; 3Colorado Division of Parks and Wildlife,Wildlife Health Program, Fort Collins, Colorado, USA

CONTACT Natália C. Ferreira [email protected]

ABSTRACT

Chronic wasting disease (CWD) is a fatal prion dis-ease which affects cervids. This disease has an asymp-tomatic incubation time of 2–4 years, and during thisperiod they can shed prions through saliva, feces,urine and placental tissue, contaminating the envir-onment. Indeed, CWD highly contagious betweencervids and shedding from live, infected animalslikely contributes to its rapid spread. So far, CWDhas been reported at least in 24 states in the US, aswell as Canada, South Korea and Norway. To date,there is no evidence of CWD transmission tohumans. However, in infected areas, deer populationcan drop as much as ~25 percent. Currently, there

PRION 139

are two tests approved to diagnose CWD: immuno-histochemistry and ELISA. However, these tests areapplied postmortem and tissue types approved are themedial retropharyngeal lymph nodes and a specificregion of brainstem (obex). Here we describe ourefforts to adapt the real time quaking-induced con-version (RT-QuIC) as a diagnostic tool to detectPrPCWD in deer ear skin. Our initial analysis of ablinded panel of 50 samples yielded 82% sensitivityand 75% specificity. We are working to improve theconditions and performance of this assay, given thatit might be useful for antemortem CWD diagnosticsand surveillance.

253. Structural bioinformatics study onmechanisms of PrPc conformational stabilizationmediated by the β 2- α 2 loop

Patricia Sotoa, Frances Mordenb,c, India Claflinc andAlyssa Bursottb,c

aPhysics department; bNeuroscience program; cBiology department,Creighton University, Omaha, NE, USA

ABSTRACT

Understanding the factors that stabilize the globular C-terminal of the cellular form of the prion protein is key todecipher PrPC to PrPSc conversion. The mechanism of thisprocess is not well understood; however, studies suggestseveral factors affect the likelihood for prion infection. Tounderstand the conformational stability of PrPC, we exam-ined the role of residue mutations, dimerization, and gpi-anchoring using structural bioinformatics techniques.Using structural bioinformatics techniques, we examinedthe role of point mutations and dimerization in the varyingsusceptibilities of PrPC structures. Our study on knownresistant sheep PrPC structures indicates that key genepolymorphisms alter the network of residue interactionsthat involve the β 2- α 2 loop. This lowered susceptibility toinitial protein misfolding likely occurs by rendering higherstructural stability than the susceptible form. Our examina-tion of prion protein dimerization suggests that the hydro-phobic interactions at the dimer interface keep the dimerstable in the bulk and on the cell surface. The rotationalmobility of dimers with respect to the cell surface is modu-lated by the gpi-anchoring state of the PrPC protein.With acomprehensive view of our results, we propose that if prionprotein fibrillogenesis occurs following the nucleation-polymerization model, then key polymorphisms raise theenergetic barrier to prevent templated prion protein con-version and dimerization conceals recognition spots thatotherwise would be recognized by PrPSc to bind and initiateprion protein conversion.

254. Humanized recombinant antibody fragmentsas tools for structural analyses of BSE and otherprions

Vineet Rathoda,b, Jimmy Lua,b, José M Flores-Fernándeza,b, Xinli Tanga,b, Razieh Kamali-Jamila,b,Leonardo Corteza,c, Assunta Senatored, SimoneHornemannd, Valerie Sima,c, Adriano Aguzzid andHolger Willea,b

aCenter for Prions and Protein Folding Diseases, University ofAlberta, Edmonton, Canada; bDepartment of Biochemistry,University of Alberta, Edmonton, Canada; cDepartment ofMedicine, University of Alberta, Edmonton, Canada; dInstitute ofNeuropathology, University of Zürich, Zürich, Switzerland

ABSTRACT

Prion diseases are rare, inexorably progressive, and fatalneurodegenerative disorders, with no therapy other thanpalliation. Prion diseases are unique compared in that theyare transmissible, whereby the infectious agent (PrPSc) ispropagated through the reproduction of its secondary,tertiary, and quaternary structure [1]. PrPSc is a conforma-tionally altered isoform of the cellular prion protein, PrPC,which is a normal cell surface glycoprotein. Prion diseasesaffect humans (e.g. Creutzfeldt-Jakob disease: CJD), as wellas a variety of animals, including chronic wasting disease(CWD) in deer and elk, scrapie in sheep and goats. BovineSpongiform Encephalopathy (BSE) is an acquired trans-missible prion disease in cattle, colloquially named ‘madcow’ disease. It was first described in 1986 in the UK andhas raised serious concerns about its transmission tohumans, where it is known as variant CJD through con-sumption of contaminated meat [2]. While the structure ofrecombinant PrPC is extensively studied using variousbiophysical techniques, no high-resolution structure ofPrPSc has been published, and only its overall architectureas a four-rung ß-solenoid is known [3,4].

Fab fragments (Fabs) are small antibody derivativesthat maintain antigen-binding capacity, and can be usedto study the structure of their antigens. One of the advan-tages of Fabs over intact antibodies is their small size,which allows them to penetrate deeper into protein aggre-gates, such as amyloid fibrils. We are using recombinantFabs as an alternate approach in deciphering the structureof PrPSc via immunogold labelling, electron microscopy,and difference mapping. One of our Fab library targetsthe four main regions of PrP: the charged cluster 1,octapeptide repeats, charged cluster 2, and globulardomain. A second set of Fabs was recombinantly engi-neered from monoclonal antibodies (mAbs) that specifi-cally recognize native PrPSc. These new PrPSc-specificmAbs (IgGs and IgMs) were recently generated in theWille lab using a rationally designed, structure-basedmimic for PrPSc.

140 ABSTRACT

The main goal is to use E. coli to express fully func-tional Fabs from both Fab libraries. Functionality assaysconducted after the expression and purification of theFabs indicate that some of them are able to bind nativePrPSc fibrils. Complexes between Fabs and purified BSEprion fibrils are being analysed using immunogold label-ling and electron microscopy. Electron micrographs ofsuitable prion fibrils will be used to generate 3D recon-structions of labelled and unlabelled BSE fibrils andprovide insights into the orientation of the respectiveepitopes in the ß-solenoid architecture of PrPSc.

References

[1] Wille H., Requena J. The structure of PrPSc prions.Pathogens. 2018;7(1):20.

[2] Aguzzi A., Sigurdson C.J. Antiprion immunotherapy:to suppress or to stimulate? Nat Rev Immunol. 2004;4(9):725–736.

[3] Requena J.R., Wille H. The structure of the infectiousprion protein and its propagation. Prog Mol BiolTransl Sci. 2017;150:341–359.

[4] Vazquez-Fernandez E., et al. The structural architec-ture of an infectious mammalian prion using electroncryomicroscopy. PLoS Pathog. 2016;12(9):e1005835.

PRION 141