* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download PDF
Synaptogenesis wikipedia , lookup
Multielectrode array wikipedia , lookup
Stimulus (physiology) wikipedia , lookup
Molecular neuroscience wikipedia , lookup
Haemodynamic response wikipedia , lookup
Nervous system network models wikipedia , lookup
Metastability in the brain wikipedia , lookup
Holonomic brain theory wikipedia , lookup
Apical dendrite wikipedia , lookup
Development of the nervous system wikipedia , lookup
Subventricular zone wikipedia , lookup
Electrophysiology wikipedia , lookup
Feature detection (nervous system) wikipedia , lookup
Clinical neurochemistry wikipedia , lookup
Optogenetics wikipedia , lookup
Neuroanatomy wikipedia , lookup
Biochemistry of Alzheimer's disease wikipedia , lookup
J Neuropathol Exp Neurol Copyright Ó 2012 by the American Association of Neuropathologists, Inc. Vol. 71, No. 5 May 2012 pp. 449Y466 ORIGINAL ARTICLE A Brain Aggregate Model Gives New Insights Into the Pathobiology and Treatment of Prion Diseases Krystyna Bajsarowicz, MS, Misol Ahn, PhD, Larry Ackerman, BA, Bernadette N. DeArmond, MD, MPH, George Carlson, PhD, and Stephen J. DeArmond, MD, PhD From the Departments of Pathology (KB, MA, BND, SJD) and Anatomy (LA), University of California, San Francisco; McLaughlin Research Institute, Great Falls, Montana (GC); and Institute for Neurodegenerative Diseases, University of California, San Francisco (SJD). Send correspondence and reprint requests to: Stephen J. DeArmond, MD, PhD, 1855 Folsom Street, Suite 505, San Francisco, CA 94143-0803; E-mail: [email protected] Krystyna Bajsarowicz and Misol Ahn contributed equally to the work. This study was supported by Grant No. NS041997 from the National Institutes of Health and by the Stephen and Patricia Schott Family Fund. CNS, including astrocytes, oligodendrocytes, and microglia. We adopted the BrnAggs model from Dr Lynn Pulliam who refined a technique first described by DeLong to generate reaggregating cell cultures from fetal mouse isocortex and hippocampus (1). Dr Pulliam used BrnAggs to study human immunodeficiency virus and herpes simplex virus encephalitis (2Y5). Ours is the first study of prion disease in BrnAggs. Brain aggregates differ from neurospheres that are composed of neural stem cells (6, 7). Two of the authors of this article (S.D. and G.C.) were coauthors of the first report of the use of neurospheres to study prion disease (8). Neurospheres are not constantly rotated, not exposed to FBS, and are often grown as monolayer cultures (9). They have been used for neural stem cell assays, as models of neurogenesis, and to expand clones of neural stem cells for transplantation purposes (10). Cultured neurospheres that express cellular prion protein (PrPC) can be infected with prions to study pathogenic prion protein (PrPSc) in a limited manner. Our earlier study showed that the susceptibility of neurosphere cultures to prions mirrored that of the parental mice and that neurospheres from Tg4053 mice, which overexpress mouse PrPC, provide a sensitive bioassay for mouse prions (8). Herva et al (11) reported that they were not able to duplicate prion inoculation of undifferentiated neurospheres but did achieve successful prion amplification in neurospheres that contained more differentiated cells. Only a few cell lines have been infectible with prions (8). The mouse neuroblastoma cell line (N2a) has been most useful for investigating the biology of prion diseases, but only a small percentage of N2a cells replicate prions when exposed to scrapie (12); moreover, prion replication in scrapie-infected N2a cells (ScN2a) has been unstable (13). Treatment of dividing ScN2a cells with quinacrine (Qa) cleared PrPSc in 5 days (14); however, Qa treatment of patients with Creutzfeldt-Jakob disease and scrapie-infected mice failed to reduce PrPSc or increase survival time. These data suggest that ScN2a cells are not a good model for drug screening (15Y18). Other prion-infectible cell lines have also shown limitations, including PC12 rat pheochromocytoma cells (19), spontaneously immortalized hamster brain cells (20), the GT1 immortalized hypothalamic neuronal cell line (21), and immortalized cells of the CNS and peripheral nervous system (22, 23). Primary cultures of neurons have also failed to support prion replication (22). The structure of BrnAggs, with their neuronal and microglial cells, resembles the organization in brain tissue. The neurons of uninfected control BrnAggs form lush dendritic trees that permit the study of prion-induced dendritic degeneration. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 449 Abstract Brain aggregates (BrnAggs) derived from fetal mouse brains contain mature neurons and glial cells. We determined that BrnAggs are consistently infected with Rocky Mountain Laboratory scrapie strain prions and produce increasing levels of the pathogenic form of the prion protein (PrPSc). Their abundant dendrites undergo degeneration shortly after prion infection. Treatment of prion-infected BrnAggs with drugs, such as a F-secretase inhibitors and quinacrine (Qa), which stop PrPSc formation and dendritic degeneration, mirrors the results from rodent studies. Because PrPSc is trafficked into lysosomes by endocytosis and autophagosomes by phagocytosis in neurons of prion strainYinfected BrnAggs, we studied the effects of drugs that modulate subcellular trafficking. Rapamycin (Rap), which activates autophagy, markedly increased light-chain 3-II (LC3-II)Ypositive autophagosomes and cathepsin DYpositive lysosomes in BrnAggs but could not eliminate the intracellular PrPSc within them. Adding Qa to Rap markedly reduced the number of LC3-IIYpositive autolysosomes. Rap + Qa created a competition between Rap increasing and Qa decreasing LC3-II. Rapamycin + Qa decreased total PrPSc by 56% compared with that of Qa alone, which reduced PrPSc by 37% relative to Rap alone. We conclude that the decrease was dominated by the ability of Qa to decrease the formation of PrPSc. Therefore, BrnAggs provide an efficient in vitro tool for screening drug therapies and studying the complex biology of prions. Key Words: Autophagosomes, Brain aggregates, Exocytosis, Prion disease, Quinacrine, Rapamycin, Scrapie. INTRODUCTION We used an in vitro brain aggregate (BrnAgg) model for the study of prion disease. The BrnAggs are made from 15-day gestational whole mouse brains grown in media supplemented with fetal bovine serum (FBS) in constantly rotated flasks. After 15 days in culture, they form spheres of 750 to 1,000 Km diameter. Brain aggregates contain mature neurons that form dendrites, axons, synapses, and all the cellular elements of the Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. Bajsarowicz et al J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 In Rocky Mountain Laboratory (RML, Hamilton, MT) scrapie prion strainYinfected BrnAggs, neurons show dendritic loss after 1 week, as seen in mice (24). Because dendritic degeneration in mice follows local PrPSc accumulation and truncation of Notch-1 to the Notch-1 intracellular domain (NICD) by the F-secretase complex (24), we explored the prevention of dendritic degeneration by treating mice with F-secretase inhibitor LY411575 (GSI) plus Qa (GSI + Qa) (17). Treatment with GSI alone produces very low NICD levels but no recovery of dendrites, which we attributed to simultaneous inhibition of F-secretase stimulators and inhibitors of dendritic growth. Treatment with GSI + Qa prevented dendritic degeneration and prevented the formation of PrPSc. Quinacrine ‘‘destabilizes’’ and fragments membrane rafts where the F-secretase complex, PrPSc, Notch-1, and other F-secretase substrates reside (25). Presumably, destabilization selectively separates some raft components from each other and permits others to interact. The GSI + Qa has its primary effect on raft domains that contain the Notch-1 inhibitor of dendritic growth and not on F-secretase substrates such as ErbB-4 and EphA4 that simulate dendritic growth (26Y28). Endocytosis of PrPSc into lysosomes and phagocytosis of PrPSc into autophagosomes are attempts to degrade and clear PrPSc from neurons. When these mechanisms fail, nerve cell dysfunction and death result. Therefore, we used BrnAggs to examine drug treatments that modify PrPSc trafficking pathways. Rapamycin (Rap) is reported to decrease PrPSc in vitro (29) and to activate autophagy by inhibiting the mammalian target of rapamycin (mTOR) (30, 31). Beneficial effects of Rap have been reported for spinocerebellar ataxia (32), Parkinson disease (33), frontotemporal lobar degeneration (34), Huntington disease (35), and amyotrophic lateral sclerosis (36). To enhance the efficacy of Rap, we added Qa (Rap + Qa), which inhibits PrPSc formation in cultured cells and in mice (14, 17). Quinacrine increases the number of lysosomes in mouse liver cells (37) but inhibits light-chain 3-II (LC3-II) turnover in mammary cells (38). Here, we used confocal microscopy and Western blot analysis for PrPSc to explore the effects of Rap alone and Rap combined with Qa on RML-infected BrnAggs. Millipore, Billerica, MA), A-3-tubulin (AB9354; Millipore), LC3II N-terminal epitope, APG8B (AP1802a; Abgent, San Diego, CA), Iba-1 (019-19741; Wako, Richmond, VA), cathepsin-D ([Cat-D] Sc 6486; Santa Cruz Biotechnology, Santa Cruz, CA), glial fibrillary acidic protein ([GFAP] 20334; Dako, Carpinteria, CA), and NeuN (MAB377, Millipore). Antibodies to PrP included R2, D18, D13, and HumP were gifts from Dr Stanley Prusiner, University of California, San Francisco. We used the following secondary antibodies for immunohistochemistry: donkey anti-rabbit IgG (711-175-152), donkey anti-human IgG (709-295-149), donkey anti-goat IgG (705-485-003), and donkey anti-chicken IgG (703-295-155). The above secondary antibodies are fluorophore conjugated and were purchased from Jackson ImmunoResearch Laboratories, Inc (West Grove, PA). For dot blot analysis, we used antibodies to PrP and biotinylated goat anti-human J chain (BA-3060; Vector Laboratories, Burlingame, CA) followed by streptavidinYhorseradish peroxidase (P0397; Dako). For nuclear staining, we used Hoechst 33342 (Invitrogen, Carlsbad, CA). MATERIALS AND METHODS Animals Pregnant wild-type FVB and FVB PrP knockout (Prnpo/o) mice were used to make BrnAgg cultures. Adult CD1 mice that have been inoculated intrathalamically with the RML strain of mouse prions were used for additional studies of the brain. All experiments were carried out in accordance with the Institutional Animal Care and Use Committee/Laboratory Animal Research Center protocol of the University of California, San Francisco. Reagents Drugs used in this study included Rap (R-5000; LC Laboratories, Woburn, MA); quinacrine (69-05-6; Sigma, St Louis, MO); F-secretase inhibitor LY411575 (gift from Drs Todd Golde and Pritam Das, Mayo Clinic, Jacksonville, FL), sodium butyrate (B5887, Sigma-Aldrich). Primary antibodies included were microtubule-associated protein 2 ([MAP-2] AB5622; 450 Preparation of BrnAgg Cultures Brain aggregates were prepared from embryonic day 15 gestation embryos from pregnant wild-type and Prnpo/o FVB mice. Brain cells were dissociated through 2 nylon meshes from a pool of embryonic brains. After 2 washes with Dulbecco’s modified Eagle medium (D-MEM H21) (UCSF Cell Culture Facility, San Francisco, CA) containing glucose (12 g/L), fungizone (2.5 mg/L), and gentamicin (50 mg/L), the dissociated neural cells were resuspended in growth medium D-MEM H21 supplemented with glucose (6 g/L), gentamicin (50 mg/L), fungizone (2.5 mg/L), and 10% FBS at a density of 1 107 cells/mL. Four milliliters of cells were placed in 25-mL DeLong flasks at constant rotation (37-C, 10% CO2). The next day, 1 mL of exchange medium D-MEM H21 supplemented with glucose (6 g/L), gentamicin (50 mg/L), and 15% FBS was added to the flask. After 2 to 3 days, the rotating BrnAggs were transferred to 50-mL DeLong flasks to which 5 mL of exchange medium was added for a total of 10 mL. Exchange media were refreshed every 2 to 3 days by removing 5 mL of conditioned medium and replacing it with 5 mL of fresh exchange medium. At 6 to 8 days, the growing BrnAggs were transferred from rotating flasks to a 24-well culture plate, and 1 to 2 BrnAggs per well were placed in 500 KL of exchange medium and rotated constantly. The medium was changed every 2 to 3 days by discarding 500 KL of conditioned medium and replacing an equal volume of exchange medium. Approximately 15 BrnAggs were obtained from the initial 4 107 neural cells. At 15 days in culture, BrnAggs were exposed for 10 days to a 1:50 dilution of RML prions derived from RML-infected CD1 mouse brains. Drug treatments began at day 25 and continued for 10 or more days. We routinely use the aggregates for 45 days. After 50 days, necrosis develops in the center of aggregates, and they are discarded. Drug Treatments Brain aggregates infected with RML prions were treated with 2.5 KM of Qa, 0.5 KM of Rap, 5.0 KM of GSI, or the combination of GSI + Qa or Rap + Qa for 10 or more days. Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Prion-Infected Brain Aggregate Model When using drugs in combination, we used the same concentration of each drug as when they were applied alone. At the end of treatment, BrnAggs were washed with PBS, harvested, and either lysed with lysis buffer (100 mM of Tris-HCl, pH 8.0; 150 mM of NaCl 0.05% Np-40, 0.5% of Na deoxycholate) and stored at j80-C for Western blot analyses or fixed with 4% formaldehyde for immunostaining. groups was quantified with the BioQuant Life Science software (Bioquant Image Analysis Corporation, Nashville, TN). Phosphotungstic Acid Precipitation of PrPC and PrPSc Fifteen BrnAggs were pooled and sonicated in lysis buffer for each of 2 uninfected control BrnAggs, 2 RML-infected BrnAggs, and 2 RML-infected BrnAggs treated with GSI + Qa groups. Total protein was measured by bicinchoninic acid assay (Pierce, Rockford, IL), and 20 Kg of protein was loaded per well on a Novex NuPAGE 4% to 12% Bis-Tris gel for total PrP. For phosphotungstic acid (PTA) precipitation, 50 Kg of lysate protein was incubated with 0.8% PTA in 2% sarkosyl/PBS buffer with complete protease inhibitor cocktails (Roche, Indianapolis, IN) for 2 hours at 37-C with continuous shaking at 350 rpm. Samples were centrifuged at 14,000 rpm for 1 hour. The entire supernatant (PTA Sup) was collected and precipitated with methanol at j20-C overnight. The methanol-precipitated PTA Sup samples were centrifuged at 14,000 rpm for 1 hour, and the PTA Sup pellets were collected and resuspended in 15 KL of lysis buffer. After adding 4 SDS sample buffer and reducing agent (Invitrogen), the samples were loaded on the gel and tested for PrPC. For PrPSc, the PTA pellet was washed with 0.2% sarkosyl/PBS, and samples were centrifuged at 14,000 rpm for 1 hour. The collected PTA pellets were resuspended in lysis buffer, and 4 SDS sample buffer with a reducing reagent was added. Samples were run on gel and transferred to a polyvinylidene fluoride membrane using iBlot (Invitrogen). After blocking with 5% milk for 30 minutes, the polyvinylidene fluoride membrane was incubated with the PrP-specific D13Y horseradish peroxidase antibody overnight. The next day, the membrane was washed with Tris-buffered salineYTween buffer for 5 minutes (3) and developed with an enhanced chemiluminescence plus Western blotting detection reagent (Pierce). For studies in mice, the neocortex was collected from uninfected and scrapie-sick CD-1 mice and homogenized using a Percellys bead beater (Percellys, Basking Ridge, NJ). One hundred micrograms of protein was used for PTA precipitation, and Western blot analysis was performed as described above. Drug Treatment of Stationary CL3.ScN2a Cells CL3.ScN2a cells were maintained at 37-C in minimum essential media (Invitrogen) supplemented with 5% FBS, 1% GlutaMax (Invitrogen), and fed with fresh media every 2 days. CL3.ScN2a cells that were approximately 70% confluent were plated and treated with Rap (1 KM) or Rap + Qa (1 KM each) in the presence of 10 mM sodium butyrate. The cells were washed with PBS, harvested, and lysed, with lysis buffer daily for 7 days. Protein concentrations were measured with bicinchoninic acid assay, and 100 Kg of protein was digested with protease K (PK) at 37-C for 1 hour. The PK-digested pellets were collected for Western blot analysis. The level of PKresistant PrPSc in untreated, Rap-treated, and Rap + QaYtreated Dot Blot Analysis of PrPSc Single BrnAggs were washed with PBS, immersed in lysis buffer, sonicated, and exposed to PK to digest PrPC. The remaining PK-sensitive proteins were precipitated with methanol in j20-C overnight. After centrifugation at 14,000 rpm for 20 minutes, the methanol-precipitated pellet was washed with PBS and reconstituted with lysis buffer. Serial 2-fold dilutions were applied to nitrocellulose membranes where PrPSc was denatured with 3M guanidinium isothiocyanate and detected with D18 antibody, followed by biotinylated goat anti-human IgG and streptavidinYhorseradish peroxidase. Horseradish peroxidase was revealed by enhanced chemiluminescence. The concentration of PrPSc in the dot blots was estimated by densitometry. Measurement of Dendritic Degeneration We used 2 methods to estimate the lengths of dendrites that had been stained with antiYMAP-2 antibodies. In the first method, 3-dimensional images of dendrites selected at random were obtained with a Zeiss LSM 510 confocal microscope that captured stacks of 1-Km-thick serial sections. To avoid detection of MAP-2Ystained cell bodies, we traced dendrites on acetate membranes, and BioQuant software was used to estimate the total dendritic load. To normalize the dendritic measurements, the density of dendrites per 100 Km2 was divided by the total number of confocal images obtained at one site and multiplied by the number of slices containing more than 3% dendrites. In the second method, lengths of dendrites were obtained from 3-dimensional images obtained with a 63 objective lens. The images were reconstructed using the IMARIS 3.0 software (Bitplane AG, Zurich, Switzerland), and the total length of dendrites was generated using the Filament Tracer module. Three sites were chosen for the highest density of dendrites in each BrnAgg, and 3 stacks of approximately 35 to 50 sections each were collected from each BrnAgg. Immunocytochemistry for Confocal Microscopy The BrnAggs were fixed in 4% formaldehyde followed by 3 washes with PBS. For detection of LC3-II, Cat-D, PrP, and Iba-1, the BrnAggs were autoclaved (10 minutes at 121-C in 0.01 M Na citrate buffer, pH 6.0), washed with PBS containing 0.3% Triton X-100 and 0.1% Tween 20, blocked overnight with 2% bovine serum albumin (BSA) and 10% normal donkey serum (NDS) and then incubated for 3 days at 4-C with antibodies specific for autophagy (LC3-II, diluted 1:75), lysosomes (Cat-D, diluted 1:100), prions (R2 and HumP, diluted 1:250), and microglia (Iba1, diluted 1:250). After incubation with primary antibodies, BrnAggs were washed with PBS containing 0.3% Triton X-100 and 0.1% Tween 20 and incubated with fluorophore-conjugated anti-human IgG (1:800), anti-goat IgG (1:800), and anti-rabbit IgG (1:800) at room temperature (RT) for 12 hours. All antibodies were diluted in PBS with 0.3% Triton X-100, 0.1% Tween 20 containing 2% BSA, and 10% NDS. For detection of dendrites, axons, and astrocytes, the BrnAggs were washed with PBS and blocked with PBS containing 0.3% Triton X-100, 2% BSA, and 10% Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. 451 Bajsarowicz et al J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 NDS for 90 minutes at RT. Primary antibodies, antiYMAP-2 for dendrites (1:1000), antiYA-3-tubulin for axons (1:100), and anti-GFAP for astrocytes (1:250) were diluted into blocking buffer and incubated for 3 days at 4-C. After 3 washes with PBS, the fluorophore-conjugated secondary antibodies corresponding to the primary antibodies were added and incubated 452 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Prion-Infected Brain Aggregate Model for 12 hours at RT. All antibodies were diluted into PBS-0.3% Triton X-100 containing 2% BSA and 10% NDS. To identify nuclei, the BrnAggs were incubated for 30 minutes at RT with Hoechst 33324 (1:5000 in PBS). Three to 4 stacks of approximately forty 1-Km-thick serial sections were captured by 2 confocal microscopes at different times (Zeiss LSM 510, Jena, Germany, or Leica Microsystems, Wetzlar, Germany). inconsistent. Equal amounts of protein were loaded on the polyacrylamide gels. No normal or abnormal PrP signal was found in the homogenates of BrnAggs derived from Prnpo/o knockout mice whether they had been exposed to RML prions (Fig. 1D, lanes 1Y3). A single band of PrPC was present in uninfected control aggregates (Fig. 1D, lane 4). The PrPSc bands corresponding to variations of the 2 N-linked glycosylation appeared in RML-infected aggregates (Fig. 1D, lane 5). Treatment of the RML-infected BrnAggs with GSI + Qa eliminated the PrPSc signal (Fig. 1D, lane 6). The absence of any detectable PrPSc in BrnAggs made from Prnpo/o mice after exposure to RML prions argues that any residual PrPSc introduced during the infection of the aggregates remained below detection or was eliminated from BrnAgg with changes of media. The sensitivity of PrPSc detection was increased with PTA precipitation of the homogenates, which selectively separate PrPSc from PrPC (41, 42). For these experiments, 90 BrnAggs were separated into 3 groups of 30 aggregates per group: uninfected controls, RML infected, and RML infected treated with GSI + Qa. Each group was further divided into 2 pools of 15 BrnAggs each for homogenization. The pool was analyzed for total PrP (PrPC and PrPSc) in the lysate, PrPC in the PTA supernatant, and PrPSc in the PTA pellet. None of the samples were digested with PK. The BrnAgg lysates showed a single PrPC band in uninfected control aggregates (Fig. 1E, Lysate, 1a,b). Three major PrPSc bands at 34, 29, and 25 kDa and a minor band at 20 kDa were seen in RML-infected aggregates (Fig. 1E, Lysate, 2a,b). A single band located at 34 kDa, which represents PrPC, was seen in RML-infected aggregates treated with GSI + Qa for 12 days (Fig. 1E, Lysate, 3a,b). In the PTA supernatant, only a single PrPC band was seen in all groups (Fig. 1E, PTA Sup). In the PTA pellet, no PrPC was identified in the control as expected (Fig. 1E, PTA Pellet, 1a,b). A very intense PrPSc signal was revealed in the untreated RML-infected PTA pellet (Fig. 1E, PTA Pellet 2a, 2b). Treatment with GSI + Qa completely eliminated conversion of PrPC to PrPSc (Fig. 1E, PTA, Pellet 3a, 3b). The intensity of the PrPC bands in RML-infected BrnAggs (Fig. 1E, PTA Sup [PrPC] 2a,b) was decreased by approximately 70% compared with uninfected controls, as measured RESULTS Growth Kinetics of BrnAggs When constantly rotated in a culture flask 15-day gestational embryonic brain cells that formed spherical aggregates from 50 to 170 Km in diameter within 72 hours (Fig. 1A; 3 days). The outer surface of the BrnAggs contained a layer of GFAPimmunopositive astrocytes (red), with their processes projecting into the interior of the aggregates. Neurons stained positively with the neuron-specific antibody NeuN (green) filled the interior of the aggregates. Brain aggregates grow by accretion of clusters of individual neural cells and by fusion with other BrnAggs (Fig. 1A; 10 days). On the sixth to eighth day of culture, BrnAggs reached 200 to 700 Km in diameter and were transferred from the flasks to constantly rotated 24-well plates. By 15 days in culture, many of the BrnAggs were as large as 1,000 Km (Fig. 1A; 15 days). Measurement of PrPC and PrPSc in BrnAggs For dot blot analysis, 3 BrnAggs exposed to RML scrapie prion strain on day 15 in culture were chosen for detection of PrPSc at 5, 20, and 35 days postinfection (dpi) (Fig. 1B). Serial dilutions of single BrnAgg lysates were treated with PK (40 Kg) to eliminate PrPC and preserve PrPSc and dotted onto nitrocellulose membranes. Densitometry of the 1:4 dilution showed that levels of PrPSc increased each dpi, as they do in scrapieinfected mice (39, 40) (Fig. 1C). For Western blot analysis, BrnAggs exposed to RML scrapie prions were homogenized in lysis buffer. The samples were not treated with PK for this analysis. Fifteen pooled BrnAggs consistently yielded detectable levels of PrPSc, whereas PrPSc signals from 5 and 10 pooled aggregates were weak and FIGURE 1. Growth characteristics of brain aggregates (BrnAggs) and measurements of pathogenic form of the prion protein (PrPSc). (A) Brain aggregates grow by progressive accretion of single cells and smaller aggregates. A confocal image immunostained for glial fibrillary acidic protein ([GFAP], red) and NeuN (green) shows the location of astrocytes and neurons. In the left panel at 3 days in culture, scale bar = 30 Km. In the right panel at 15 days in culture, scale bar = 100 Km, and applies also to the middle panel from day 10. (B) The PrPSc is detected by dot blot analysis of 3 single aggregates; 4 dilutions from 3 time points in culture are shown. (C) The mean and SD of the 1:4 dilution of PrPSc were measured by densitometry (arbitrary units). (D) Western blot analysis was used to measure cellular prion protein (PrPC) and PrPSc in the pools of 15 BrnAggs made from PrP knockout (Prnpo/o) (lanes 1Y3) and wild-type FVB mouse embryos (lanes 4Y6). Neither PrPC nor PrPSc is detected in the Prnpo/o samples. In BrnAggs derived from FVB embryos, PrPC is detected in uninfected BrnAggs (lane 4); PrPSc is detected in aggregates exposed to Rocky Mountain Laboratory (RML) scrapie prion strain prions (lane 5); little PrPSc is detected in BrnAggs exposed to RML prions and treated with F-secretase inhibitor (GSI) quinacrine (Qa) for 10 days (lane 6). (E) Phosphotungstic acid (PTA) precipitation of homogenates from 2 pools of 15 BrnAggs yields characteristic the PrPC and PrPSc bands after 35 days in culture. Lanes 1a and 1b are uninfected BrnAggs; lanes 2a and 2b are BrnAggs exposed to RML prions on day 15 in culture; lanes 3a and 3b are BrnAggs infected with RML prions at 15 days and treated with GSI + Qa for 10 days starting at day 25. (F) Phosphotungstic acid precipitation of CD-1 mouse neocortex uninfected (lanes 1Y3) and infected with RML prions (lanes 4Y6); mice were killed at 130 days postinfection (dpi). The conversion of PrPC to PrPSc reduces PrPC. Intense PrPC bands are found in PTA supernatant (S) in the neocortex of the 3 uninfected control mice. The intensity of the PrPC bands (S) is weaker when the PrPSc is abundant in the PTA pellet (P). (G) Densitometry shows that PrPC was significantly reduced by 95% when PrPSc is formed. **t-test probability of a decrease p G 0.01. Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. 453 Bajsarowicz et al J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 by densitometry (data not shown). The decrease was associated with the intense PrPSc bands in RML-infected BrnAgg (Fig. 1E, PTA Pellet, 2a,b). Such a decrease in PrPC was consistent with data showing a 95% depletion of PrPC in RML- infected CD-1 mice, which was caused by the rapid conversion of PrPC to PrPSc (Figs. 1F, G). After GSI + Qa treatment of RML-infected BrnAggs, PrPC levels were approximately 58% lower than in uninfected FIGURE 2. (AYC) The number of microtubule-associated protein 2 (MAP-2)Ypositive dendrites per given volume (dendritic density) was estimated in 3 uninfected (AYC) and 3 Rocky Mountain Laboratory (RML) scrapie prion strainYinfected brain aggregates (BrnAggs) (D, E). (F) To estimate dendrite densities, MAP-2Ystained dendrites in each confocal slice were drawn onto acetate membranes, and the density in each slice was measured by BioQuant morphometry. **Probability of a difference between control and RML-infected BrnAggs at 25 days, p = 0.028; at 35 days, p = 0.032 (n = 3 BrnAggs each). (G) The responses of dendritic lengths to 10-day treatments with F-secretase inhibitor (GSI), quinacrine (Qa) and GSI + Qa were measured. Control represents uninfected BrnAggs. RML (UnTx), RML-infected BrnAggs without treatment. The lengths of dendrites immunostained with MAP-2 were measured with IMARIS 3 software using confocal microscopy to generate a 3-dimensional image. n = number of BrnAggs analyzed. (H) Immunocytochemistry for pathogenic form of the prion protein (PrPSc, red) and cathepsin-D (Cat-D, green) in a BrnAgg neuron early in RML infection (10 days postinfection) showing prominent plasma membrane immunostaining for PrPSc and Cat-DYimmunopositive lysosomes with a minimum number of merged PrPSc (yellow). Nuclei are stained with Hoechst (blue). Scale bars = (E) 30 Km; (AYD, H) 10 Km. 454 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Prion-Infected Brain Aggregate Model controls, which indicates that the GSI + Qa treatment did not restore PrPC levels, although it reduced PrPSc levels by 100% (Fig. 1E, PTA Lysate 3a, 3b). dritic degeneration was detectable approximately 1.5 weeks after PrPSc began to form in the brain region. When BrnAggs were infected with RML prions on culture day 15, degeneration of dendrites was detectable within 5 days after PrPSc accumulation. To measure dendritic densities, dendrites were immunostained by MAP-2 and traced with a felt-tipped pen on an acetate membrane on the computer screen. The drawings were quantified with BioQuant Morphometry software to estimate dendritic loads. Three 40 confocal stacks were obtained from 3 to 4 BrnAggs at 15, 25, and 35 days in culture. In uninfected control BrnAggs, the density of dendrites doubled from 15 to 25 days (Figs. 2A, B). At 35 days, the density of dendrites decreased to the 15-day level presumably as a result of pruning back of the dendrites with maturation (Fig. 2C). In contrast, BrnAggs infected with RML prions showed a progressive decrease in the length and number of dendrites at 25 and 35 days (Figs. 2D, E). BioQuant measurement of dendritic densities showed a significant decrease in RMLinfected BrnAggs at 25 and 35 days (10 and 20 dpi) versus uninfected controls (p G 0.03, n = 3) (Fig. 2F). Dendritic Degeneration in Prion-Infected BrnAggs Among the earliest neurodegenerative changes seen in experimental RML scrapie are shortening of dendrites and a decrease in the number of dendritic branches (24). In mice and BrnAggs, dendritic degeneration occurs in response to PrPSc accumulation in plasma membranes (Fig. 2H) (43). Therefore, we measured the growth rate of dendritic density in uninfected control and in RML-infected BrnAggs. In mouse studies, den- Dendritic Loss Is Not Caused by Loss of Neurons We evaluated the MAP-2 staining as a function of the number of neurons at 20 dpi to RML prions (35 days in culture) and compared them with uninfected cultures. Differentiated neurons were identified by MAP-2 staining of their cytoplasm (44). In 3 uninfected cultures, nerve cell bodies with multiple attached dendrites showed intense perinuclear MAP-2 staining and deposition of MAP-2 throughout the remainder of the enlarged nerve cell body (Fig. 3A). In contrast, 3 RMLinfected BrnAggs showed very few MAP-2Ypositive dendrites and lack of enlargement of neuronal cell bodies, but they did show prominent perinuclear MAP-2 staining (Fig. 1B, arrows). We counted all neurons with perinuclear MAP-2 in each confocal stack whether or not they had attached dendrites. The RML-infected BrnAggs had 30% to 40% more neurons fulfilling the minimum MAP-2 criteria (Fig. 1C). This increase in the numbers of neurons was significant (p G 0.03, n = 3). These data indicate that dendritic degeneration occurred without loss FIGURE 3. Degeneration of dendrites during scrapie infection is not associated with a decrease in the number of neurons. Dendrites and nerve cell bodies are stained with microtubuleassociated protein 2 (MAP-2, red). Nuclei are stained with Hoechst (blue). (A) Representative of 3 uninfected brain aggregates (BrnAggs) at 35 days in culture showing many neurons with long and branched dendrites, dendrite-bearing neurons that have enlarged MAP-2Ypositive cell bodies, and neurons without dendrites that have perinuclear MAP-2 staining. (B) Brain aggregates at 35 days in culture (n = 3) that were infected with Rocky Mountain Laboratory (RML) scrapie prion strain prions for 20 days contain a few neurons with short and unbranched dendrites, atrophied cell bodies, and many neurons with perinuclear MAP-2 staining. Arrows indicate neurons without dendrites but with perinuclear MAP-2 staining. (C) Number of all neurons identified with perinuclear MAP-2 staining are significantly increased in RML-infected BrnAggs versus control (p G 0.05 by t-test). Analysis is based on stacks of 1-Km-thick confocal images. Scale bar = (A, B) 30 Km. Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. 455 Bajsarowicz et al J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 of neurons. This conclusion is consistent with the fact that dendritic loss in prion disease is caused by PrPSc activation of Notch-1 and can be prevented by treatment of mice and BrnAgg with GSI + Qa. One possibility that more neurons displayed MAP-2 staining may be caused by the existence of neuronal stem cells in the BrnAggs that do not express MAP-2. Another possibility is that to rebuild dendrites lost because of RML infection, neurons released dendritic growth factors from presynaptic nerve endings that act on all neurons to induce MAP-2. FIGURE 4. Confocal 1-Km-thick images of uninfected brain aggregates (BrnAggs). (A, B) Light-chain 3-II (LC3-II, red), cathepsin-D (Cat-D, green) and Hoechst stain (blue) at 25 (A) and 37 (B) days in culture are shown. Yellow color indicates colocalization of LC3-II and Cat-D. The rectangle in (A) corresponds to the images in (C) and (D). The inside of the BrnAggs are marked with an ‘‘i’’ and outside the BrnAggs with an ‘‘o.’’ (C) Higher magnification of the image in (A) shows that LC3-II (red) localized in neuronal nuclei (lavender color), in the nerve cell body (oval), and in the neuropil away from the cell body (arrows). (D) Same field as in (C) but stained for Cat-D (green). Cell body lysosomes are distributed in numbers, and distribution is similar to that in Figure 2H. Scale bars = (A, B) 30 Km; (C, D) 20 Km. 456 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Prevention of Dendritic Degeneration Given that dendrites degenerate in BrnAggs as they do in vivo, we tested whether treatment with GSI, Qa, or GSI + Qa would prevent dendritic degeneration as in mice (17). The effects of each drug treatment were evaluated on 4 to 7 BrnAggs. The 3 sites chosen in each BrnAgg contained the highest density of dendrites. Treatment with GSI (5 KM), Qa (2.5 KM), or GSI + Qa (same doses) was begun 10 days after the BrnAggs had been exposed to RML prions and continued for 10 more days. Mean total lengths of dendrites and standard deviations were measured (Fig. 2G). Untreated RML infection reduced the lengths of dendrites by approximately 40% compared with the lengths in uninfected control BrnAggs. Treatment with GSI had virtually no effect on dendritic lengths. Treatment with Qa preserved dendritic lengths 75% compared with those of controls. In the GSI + QaYtreated BrnAggs, dendritic lengths were similar to those in uninfected controls. Thus, GSI + Qa effects on PrPSc levels (Figs. 1D, E) and on dendrites verify that the BrnAgg model accurately reproduces treatment effects in mice. Confocal Microscopy We used confocal microscopy in most to localize PrPSc, Cat-DYimmunopositive lysosomes, and LC3-IIYimmunopositive autophagosomes in groups of 3 to 5 uninfected control, RMLinfected untreated, and RML-infected and Rap-, Qa-, or Rap + QaYtreated BrnAggs. In each of these categories, the 3 to 5 BrnAggs yielded different but characteristic distributions of PrPSc, lysosomes, and autophagosomes. Representative confocal images emphasize the characteristic similarities and differences, although the confocal data are not quantitative. Subcellular Compartments in Uninfected BrnAggs At day 25 in culture, many nuclei in uninfected control BrnAggs exhibited LC3-II (red) immunoreactivity that combines with Hoechst (blue) to produce a lavender color (Figs. 4A, C). At this time point, punctate deposits of LC3-II filled the neuronal cell body (Fig. 4C) and most of the neuropil between nerve cell bodies (Fig. 4C, arrows). A nuclear form of LC3 that regulates levels of fibronectin mRNA has been described (45). The presence of soluble LC3 in the nucleus or cytoplasm becomes less obvious as levels of LC3 become associated with autophagosomes. LC3 was originally identified in the cytoplasm as a microtubule-associated protein (46, 47). LC3-II interacts with the dendrite-specific Ca2+-sensing protein, caldendrin (48). These latter 2 associations indicate that some of the cell body immunostaining and the neuropil immunostaining of LC3-II were within dendrites and were likely participating in the pruning back of dendrites between 25 and 35 days in culture (Figs. 2AYC, F). Large numbers of lysosomes in the nerve cell bodies at 25 days were also probably contributing to pruning back the dendritic trees (Fig. 4D). At 37 days, the number of LC3-II particles and Cat-D-immunopositive lysosomes were greatly diminished. In neurons of uninfected BrnAggs at 25 days in culture, lysosomes filled the neuronal cytoplasm and tend to surround their nuclei (Fig. 4D). Small numbers of LC3-II autophagosomes were scattered among the lysosomes, and some of the lysosomes and autophagosomes were merged (Fig. 4A, yellow). At day 37 in culture, most of the LC3-II had migrated out of nuclei and appeared to be associated with autophagosomes Prion-Infected Brain Aggregate Model based on the size of the LC3-II particles and their subcellular location scattered among lysosomes (Fig. 3B). Other LC3-II deposits were scattered in the neuropil. Very few LC3-II particles appeared merged with Cat-D (Fig. 4B). PrPSc in Subcellular Compartments The temporal stages of subcellular PrPSc accumulation in BrnAggs infected with RML prions were similar to those found in vivo (43, 49). In the rodent brain, PrPC was converted to PrPSc in the plasma membrane and/or in early endosomes (50). Accumulation of PrPSc in the neuronal plasma membrane occurred rapidly and caused a dysfunction of membrane components followed by degeneration of the plasma membrane and synaptic structures (51, 52). In the following 3 to 6 weeks, PrPSc accumulated in sufficient amounts in lysosomes and autophagic vacuoles, where it triggered further nerve cell dysfunction and, with the help of microglia, nerve cell death (49, 53, 54). At 5 and 10 dpi, PrPSc was confined to the neuronal plasma membrane in some RML-infected BrnAggs (Fig. 2H). By 22 dpi, PrPSc colocalized with the Cat-DYimmunopositive (green) lysosomal compartment (Figs. 5A, C). Many of the remaining small and large aggregates of PrPSc in the neuronal cell body were adjacent to the lysosomal compartment (Fig. 5C). These intracellular aggregates of PrPSc colocalized with LC3-IIYimmunopositive autophagosome (yellow) (Figs. 5B, D). Compared with the 37-day control (Fig. 4B), there was no or only a small increase in the number of lysosomes; however, the RML-infected lysosomes at 22 dpi (37 days in culture) tended to distribute abnormally in the nerve cell body, where they formed adherent clusters, in which some contained PrPSc and displayed yellow colocalization with green Cat-D (Fig. 5C). The PrPSc bearing Cat-D and LC3-II compartments did not colocalize (data not shown). In RML-infected BrnAggs, LC3-II staining was also seen in a large number of particles, 1 to 4 Km in diameter, scattered throughout the neuropil and overlying some neuronal nuclei (Figs. 5B, D, green). A few of these particles were seen in the uninfected control BrnAggs at 37 days in culture (Fig. 4B). One possibility is that they were populations of autophagosomes that accumulated in axons and presynaptic boutons during prion disease, as we will discuss below (52, 55). Rap Effects Rapamycin has been shown to decrease PrPSc in vitro (29, 56) and to increase autophagy (30, 31). Rapamycin, also known as sirolimus, has manageable side effects and has been approved for chronic treatment in humans to prevent rejection after renal transplantation (Prescribing Information for Rapamune [sirolimus], Pfizer, Inc, 2010). We tested the ability of Rap and Rap + Qa to clear PrPSc from CL3.ScN2a cells, in which cell division had been arrested with sodium butyrate. Rapamycin alone did not clear PrPSc from CL3.ScN2a cells (Fig. 6). Treatment with Rap alone was identical to untreated CL3.ScN2a cells. In contrast, Rap + Qa rapidly cleared PrPSc in 1 to 2 days and held the markedly reduced PrPSc levels for another 5 days (Fig. 6). Therefore, we used BrnAggs to explore the effect of Rap or Rap + Qa on PrPSc levels. Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. 457 Bajsarowicz et al 458 J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Prion-Infected Brain Aggregate Model was examined with Cat-D (green), very little intracellular LC3II was identified, and the small amount of LC3-II that was present showed little or no merging with Cat-D (Fig. 8C). Rap + Qa Effects FIGURE 6. Treatment of nondividing CL3.ScN2a cells with rapamycin (Rap) alone had no effect on the pathogenic form of the prion protein (PrPSc) levels; Rap + quinacrine (Qa) markedly reduced PrPSc levels within 2 days of treatment. The Rap alone curve is very similar to that of untreated CL3.ScN2a cells. At 20 dpi with RML prions and after 10-day treatment with Rap, BrnAggs had large increases in the number of Cat-DYimmunopositive lysosomes (Fig. 7A) and LC3-IIY immunopositive autophagosomes in neurons (Fig. 7B). Some neurons appeared full of merged Cat-D and LC3-II, suggesting that a single autolysosome compartment had been formed (Fig. 7C). The PrPSc colocalized with both the Cat-D compartment (Fig. 7D) and the LC3-II compartment (Fig. 7E) and remained, for the most part, intracellularly within neurons. Very little PrPSc accumulated at the periphery of the BrnAgg (Figs. 7Di, Ei). Rapamycin alone treatment did not appear to degrade PrPSc in lysosomes, autophagosomes, or autolysosomes, consistent with its protease resistance. Qa Effects In nondividing CL3.ScN2a cells, continuous treatment with Qa initially decreased PrPSc levels; but after 2 days, the levels rebounded (57). Quinacrine increased the number of lysosomes in mouse liver cells (37) but inhibited LC3-II turnover in mammary cells (38). Similarly, treatment of BrnAggs with Qa alone for 17 days dramatically eliminated LC3-II staining in the cell bodies of neurons (Fig. 8B) and increased the number of Cat-D lysosomes (Figs. 8A, C). It also appeared to decrease the number of the individual punctate LC3-II particles in the neuropil (Fig. 8B). The PrPSc colocalized with a few of the punctate deposits of LC3-II (Fig. 8B). In evaluating PrPSc treated with Qa in BrnAggs, panels A and B of Figure 8 show an overall decrease of PrPSc in the nerve cell body but an increase of PrPSc at the periphery of the BrnAgg. Western blot analysis showed a statistically significant 37% decrease in PrPSc (p G 0.05, n = 3) (Figs. 9A, B). When the LC3-II (red) Rapamycin + Qa treatment appeared to trigger a conflict between the effect of Rap to increase LC3-II autophagosomes (Fig. 7B) and the inhibitory effect of Qa on autophagosomes (Fig. 8B). After 12 days of treatment with Rap + Qa, Qa dominated the inhibitory effect on LC3-II. It cleared most of the LC3-II compartment from nerve cell bodies (Fig. 10B); however, the Rap effect on autophagosome left some LC3-II in the cell bodies, which merged with Cat-D lysosomes (Fig. 10C). The Cat-D lysosomes in the cell body tended to be more irregularly arranged with Rap + Qa (Fig. 10A); whereas with Qa, the lysosomes tended to be the same size and evenly dispersed in the cell body (Figs. 8A, C). Some PrPSc remained associated with Cat-D lysosomes (Fig. 10A). Other deposits of PrPSc occurred in the cell bodies of cells that did not contain Cat-D or LC3-II (Figs. 10A, B). These cells are most likely microglia (see below). With Rap + Qa, Western blot analysis showed a statistically significant 56% decrease of PrPSc (Fig. 9). Extracellular PrPSc Is Ingested by Microglia We examined the extracellular space for the presence of microglia and evaluated their capability to ingest PrPSc. Microglia identified with the Iba-1 antibody (green) displayed yellow immunolabeling after merging with PrPSc (red) (Fig. 11). The number of double-immunolabeled yellow signals varied with infection and treatment. Uninfected BrnAggs showed very few resting microglia, with a very small amount of cytoplasm containing merged PrPC and Iba-1 (Fig. 11A). Untreated RML-infected BrnAggs showed small amounts of PrPSc in the extracellular space and relatively few microglia with the merged signals (Fig. 11B). Treatment with Qa showed the least amount of merged PrPSc and Iba-1 (Fig. 11C), which indicated that Qa did not cause proliferation of activated microglia. Treatment with Rap showed an intermediate level of merged signal (Fig. 11D), indicating that larger numbers of activated microglia had ingested PrPSc. Treatment with Rap + Qa showed the largest amount of ingested PrPSc and the highest amount of merged PrPSc and Iba-1 (Fig. 11E). The increased volume of microglial cytoplasm after GSI + Qa treatment indicates that the microglia were activated. DISCUSSION Brain aggregates provided a clear view of the distribution of PrPSc in the plasma membrane, lysosomes, and autophagosomes of neurons. They also allowed us to view the release of PrPSc into the extracellular space and ingestion FIGURE 5. Confocal images of untreated, Rocky Mountain Laboratory (RML) scrapie prion strainYinfected brain aggregates (BrnAggs), 1 Km thick, 20 days postinfection (dpi). (A) Low-power view stained for pathogenic form of the prion protein (PrPSc, red), cathepsin-D (Cat-D, green) and Hoechst (blue). Colocalization of PrPSc with Cat-D is yellow. (B) Low-power view of PrPSc (red), lightchain 3-II (LC3-II, green) and colocalization of PrPSc with LC3-II (yellow). The inside of the BrnAgg and the outside culture medium are indicated by i and o, respectively. (C) Higher power of the neuron marked with an asterisk (A). The PrPSc (red) is not detected in the plasma membrane of neurons. The overlap of PrPSc and Cat-D (Green) is yellow (merge). The remaining intraneuronal PrPSc is adjacent to lysosomes (arrow). (D) Most of the PrPSc (red) adjacent to lysosomes are located in LC3-II (green)Ypositive autophagosomes (yellow) (arrow). Asterisks indicate the same neuron in all images. Scale bars = (B, D) 20 Km; ([A, C], respectively) 20 Km. Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. 459 Bajsarowicz et al 460 J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Prion-Infected Brain Aggregate Model of PrPSc by microglia. Using specific markers to identify lysosomes and autophagosomes, we monitored 3 subcellular mechanisms that control trafficking of PrPSc in neurons: endocytosis, phagocytosis, and exocytosis. Treatment with Rap alone indicated that most of the PrPSc was not degradable by cellular enzymes, meaning that PrPSc accumulated inside neurons in the BrnAgg. We also have preliminary data suggesting that PrPSc was not released from the extracellular space to the medium surrounding BrnAggs (unpublished data) and speculate that decreases in PrPSc levels must result from decreased formation of PrPC to PrPSc in membrane rafts. Neurons in BrnAggs form luxuriant dendritic trees. The response of dendritic degeneration to drug treatment during prion disease was identical in BrnAggs to the results obtained in mice in which dendritic degeneration was the result of PrPSc accumulation in plasma membranes that activated the F-secretase complex cleavage of Notch-1 to the NICD (17). Accumulation of PrPSc in plasma membrane was verified in BrnAggs at early dpi with scrapie (Fig. 2H). The NICD activates the HES and HERP families of inhibitor effecter proteins, which in turn inhibit proneuronal genes that maintain the length and branching of dendrites (58). Because GSI + Qa treatment prevented PrPSc formation and dendritic degeneration in both mice and BrnAggs, we conclude that BrnAggs accurately mimic the in vivo condition. Transport of PrPSc by endocytosis from the plasma membrane to late endosomes and lysosomes is well established (43, 49, 53, 59). In BrnAggs, subcellular trafficking of PrPSc follows the same route as in animals. Cathepsin-D immunohistochemistry demonstrated aggregation and clustering of lysosomes and colocalization with PrPSc as in scrapie-infected mice (43, 49). In BrnAggs, extralysosomal PrPSc in neuron cell bodies colocalized with LC3-IIYpositive autophagosomes. This is also consistent with data from electron microscopy that showed large aggregates of lysosomes, autophagosomes, and multivesicular bodies in the neuron cell bodies of rodents with prion disease (60). We also detected LC3-IIYpositive autophagosomes intermingled with lysosomes. The LC3-II immunoreactivity was seen as 1- to 4-Km deposits scattered throughout the neuropil, suggesting that it is located in axons during prion disease. By electron microscopy, autophagosomes with double membranes are located in axons, axon terminals, and presynaptic boutons in prion diseases (52, 55). In uninfected control BrnAggs, LC3-II immunopositivity varied with time in culture and was different from that in RML prion-infected BrnAggs. At 25 days in culture, LC3-II was located in the nuclei of many neurons, in the cell body, and in the neuropil away from the cell body. By 37 days in culture, LC3-II in the cell body becomes associated with autophagosomes, and nuclear staining is largely absent. The large amount of LC3-II in the neuropil between nerve cell bodies seen at 25 days in culture is probably not associated with autophago- somes but with microtubule-associated protein and dendritespecific Ca2+-sensing protein caldendrin in dendrites (46Y48). This correlates with the evidence that LC3-II expression is increased during dendritic growth (61). The neuropil location of LC3-II occurs at the peak of dendritic growth in uninfected BrnAggs. The pruning back of dendrites during the succeeding 10 days correlates with the decrease of LC3-II in the neuropil. Immunocolocalization of LC3-II in dendrites requires autoclaving to make the epitopes available to LC3-IIYspecific antibodies. We found, however, that autoclaving precludes detection of MAP-2Ypositive dendrites and A-3-tubulinY positive axons (unpublished data). Therefore, we were unable to verify that LC3-II was located in dendrites. Quinacrine is reported to increase the number of lysosomes in liver cells (37) and to inhibit LC3-II turnover in mammary cells (38). In addition to destabilizing membrane rats, we found that Qa exerts a broad range of effects on RML-infected BrnAggs, which increased the number of Cat-DYimmunopositive lysosomes in neural cells and caused an almost complete reduction of LC3-II in RML-infected BrnAggs. Western blot measurements indicated that Qa treatment decreased PrPSc levels by 37% versus untreated and Rap aloneYtreated infected BrnAggs (Fig. 9). Quinacrine increased the number of Cat-D lysosomes, but they do not degrade PrPSc. Very few activated microglial cells were present in the extracellular space, and they do not appear to degrade PrPSc either. Therefore, the decrease of PrPSc was most likely caused by effects of Qa on membrane rafts, which would prevent PrPC from interacting with PrPSc and forming more PrPSc. In the usual progression of autophagy in normal cells, autophagosomes with double limiting membranes bind to lysosomes to acquire acidic hydrolases and then mature into autolysosomes with a single limiting membrane (62). In BrnAggs, the discovery of PrPSc in LC3-IIYpositive autophagosomes is new evidence that this phagocytic pathway is active in prion disease as has been reported in Alzheimer disease (63, 64). Both PrPC and PrPSc are released from RMLinfected cells into the extracellular space by exocytosis (65, 66). The cellular mechanisms underlying this form of exocytosis have been uncovered in studies of lipid raft proteins, including glycophosphatidylinositol-anchored proteins such as PrPC and PrPSc (67). Developing autophagosomes presumably sequester plasma membrane fragments containing PrPC and PrPSc (68Y70). The PrPSc-containing autophagosomes fuse with the multivesicular bodies derived from the endosomal compartment, and the resulting hybrid structure binds to the plasma membrane to release its contents into the extracellular space. Normal neurons contain very few autophagosomes, whereas in prion diseases, neuron cell bodies contain large clusters of a mixture of lysosomes, autophagosomes, and multivesicular bodies that are partially fusing or fully fused (60). In prion disease, distal axons and presynaptic boutons also contain FIGURE 7. (AYE) In Rocky Mountain Laboratory (RML) scrapie prion strainYinfected brain aggregates (BrnAggs), Rapamycin (Rap) alone increases the number and amount of cathepsin-D (Cat-D, green) (A) and light-chain 3-II (LC3-II, red) (B) in neurons and merges the 2 compartments (yellow) (C). Most of the pathogenic form of the prion protein (PrPSc, red) colocalize with the Cat-D (green) compartment (yellow) (D). The PrPSc (red) also colocalize with the LC3-II compartment (green) (E). See Figure 5 for appropriate untreated control. ‘‘o’’ indicates outside the BrnAgg in (D) and (E). Scale bar = (AYE) 20 Km. Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. 461 Bajsarowicz et al J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 FIGURE 8. In Rocky Mountain Laboratory (RML) scrapie prion strainYinfected brain aggregates (BrnAggs), quinacrine (Qa) alone seems to increase the number of cathepsin-D (Cat-D, green) lysosomes (A), which was verified by merging the Cat-D signal (green) with the light-chain 3-II (LC3-II, red) (C). Quinacrine mainly eliminates most of the LC3-II signal (green), except the relatively small numbers of diffusely distributed LC3-II particles (B). Arrows in (B) indicate colocalization of punctate LC3-II particles with the pathogenic form of the prion protein (PrPSc, red). In (A), (B) and (C), inside and outside the BrnAgg are indicated by ‘‘o’’ and ‘‘i.’’ Scale bar = (AYC) 30 Km. 462 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 FIGURE 9. Groups of 15 brain aggregates (BrnAggs) were subjected to Western blot measurements of total pathogenic form of the prion protein (PrPSc). The groups were untreated (UnTx), Rapamycin (Rap) aloneYtreated, quinacrine (Qa) aloneYtreated, and Rap + QaYtreated BrnAggs. (A) Representative Western blot not exposed to proteinase K shows PrPSc and A-actin. (B) Densitometry measurements of the 3 untreated Rocky Mountain Laboratory (RML) scrapie prion strainYinfected samples were given the same value because the measurements varied considerably for the 3 groups of data. The mean and standard deviations and t-test were determined for the 3 treatments. *t-test probability (p G 0.05, n = 3) for a decrease of PrPSc for Qa alone and Rap + Qa versus Rap alone. autophagosomes and no obvious multivesicular bodies adjacent to disintegrating plasma membranes (52, 55). It is possible that some autophagosomes can release their contents including PrPSc into the extracellular space via local degenerating membranes. In untreated RML-infected BrnAggs, most of the autophagosomes did not appear to bind to lysosomes; instead, they were positioned adjacent to lysosomes and existed as large and small aggregates of PrPSc that colocalized with LC3-IIY positive autophagosomes. It has been reported that autophagosomes containing AA-42 peptide in Alzheimer disease function abnormally and may be the origin of most of the AA42 in the brain (63, 64). Autophagosomes containing PrPSc enter an ‘‘unconventional’’ exocytosis pathway that deposits PrPSc in the extracellular space (68Y70). It is assumed that protease-resistant PrPSc is not degradable by lysosomal enzymes. It is also assumed that the BrnAgg is a closed system, that is, that PrPSc formed and released by neurons stay within the BrnAgg. We need to Prion-Infected Brain Aggregate Model verify this by collecting media for 1 week from RML-infected BrnAgg cultures and assaying for PrPSc. Rapamycin alone did not produce a significant decrease of intracellular PrPSc as a result of exocytosis when compared with PrPSc levels in untreated RML-infected BrnAggs. This was attributed to storage of PrPSc intracellularly within increased numbers of Cat-D and LC3-II compartments. However, if a sizable proportion of total PrPSc were protease sensitive, a significant decrease in total PrPSc would have been expected. Treatment with Rap + Qa produced a significant decrease in PrPSc (Fig. 9). Confocal microscopy revealed competition between activation of autophagy by Rap (30, 31) and inhibition of autophagy by Qa (38) (Fig. 10). Merging of the Cat-D and LC3-II signals demonstrated a modest overlap of lysosomes and autophagosomes (Fig. 10C). Treatment with Rap alone led to a much larger overlap (Figs. 7AYC) that contained undegraded PrPSc. Western blot analysis showed an approximately 10% decrease in PrPSc levels compared with untreated controls (Fig. 9). Together, these data support the conclusion that most of the PrPSc accumulated in lysosomes, autophagosomes, and autolysosomes and was not degraded. The overlap of Cat-D lysosomes and LC3-II autophagosomes with Rap + Qa accounted for 5% to 10% decrease of PrPSc, as did with Rap alone. Therefore, Qa effects on membrane lipid rafts likely caused the remaining approximately 46% decrease in PrPSc. The LC3-II autophagosomes predictably fuse with multivesicular bodies, which lead to unknown amounts of PrPSc exocytosis. As a surrogate marker of exocytosis, we used Iba-1 antibodies to mark microglia and PrP antibodies to identify ingested PrPSc. In scrapie-infected BrnAggs, activated microglia ingested PrPSc. They do not form significant amounts of PrPSc de novo. Activated microglia with ingested PrPSc release cytokines that cause nerve cell death of neurons expressing PrPC (71). We found the highest numbers of activated microglia with the Rap + Qa treatment followed by Rap alone. We believed that Rap activates microglia, although it is reported to inhibit microglial activation (72). It is possible that the relatively large amount of PrPSc ingested by microglia caused their activation; an increased volume of the microglial cell body identifies activation. In RML-infected BrnAggs that were untreated or treated with Qa only, we observed very low numbers of activated microglia with a small amount of ingested PrPSc. Uninfected BrnAggs did not have recognizable activated microglia. Therefore, we conclude that PrPSc was transported to the extracellular space, particularly following treatment with Rap + Qa and Qa alone. The present results explain why Rap alone did not clear PrPSc from CL3.ScN2a cells (Fig. 6). Rapamycin alone increased LC3-II autolysosomes and Cat-D lysosomes in BrnAggs. The PrPSc became stored in both of those compartments, and it appeared that very little PrPSc was removed from neurons by multivesicular bodyYfacilitated exocytosis. Western blot analysis showed an approximately 10% decrease of total PrPSc versus untreated controls. In contrast, Rap + Qa treatment of CL3.ScN2a cells cleared PrPSc by 100% in 2 days. In BrnAggs, Rap + Qa reduced total PrPSc by 56%. Confocal microcopy showed that the Qa component reduced the number of LC3-II autophagosomes and merged some indigestible PrPSc with Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. 463 Bajsarowicz et al J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 FIGURE 10. (A, B) In Rocky Mountain Laboratory (RML) scrapie prion strainYinfected brain aggregates (BrnAggs), rapamycin (Rap) + quinacrine (Qa) treatment has little effect on cathepsin-D (Cat-D)Ypositive lysosomes (A) but markedly reduces the number of light-chain 3-II (LC3-II)Ypositive autophagosomes (B). Pathogenic form of the prion protein (PrPSc, red) is located in neurons where it variably merges with Cat-D (large arrows) (A) but not with LC3-II (B). The small arrows in (A) and (B) indicate cells that contain PrPSc but not merged with Cat-D or LC3-II, respectively. Most of the PrPSc appears to be beneath the outer perimeter of the BrnAggs. The region outside of the BrnAggs is marked with an ‘‘o.’’ (C) The LC3-II (red) and Cat-D (green) are merged (yellow) in some neurons from a different field than shown in (A) and (B). Scale bar = (AYC) 20 Km. 464 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 Prion-Infected Brain Aggregate Model FIGURE 11. (AYE) Pathogenic form of the prion protein (PrPSc, red) is present in the extracellular space where is ingested by microglia identified with Iba-1 (green). Merging the PrPSc and the Iba-1 stains (yellow) signifies ingestion. Scale bar = (AYE) 30 Km. Cat-DYpositive lysosomes in neurons and with Iba-1Ypositive microglia in the extracellular space. Because PrPSc is largely indigestible, Rap + Qa decreased PrPSc formation by the effect of Qa on membrane rafts. Most of the PrPSc remained in the BrnAgg within neurons, within the extracellular space, and within microglia. In CL3.ScN2a cells, we expect that PrPSc formation was reduced by the Qa component of Rap + Qa, and all PrPSc was discharged from the cells by exocytosis. Exocytosed PrPSc was removed from the culture with media change. To confirm this hypothesis, we will collect media from the cell culture to test for the presence of PrPSc. In summary, we examined the behavior of RML prions in BrnAggs in detail. Brain aggregate cultures are a suitable model because their neurons and glia interact with each other as much as they do in the CNS. The neuronal-glial relationship most likely fostered prion infection, prion propagation, and neuronal dysfunction. The results explain why Rap alone treatments of many genetic disorders and prion disease have had limited success. In our preliminary study in CD-1 mice, Rap + Qa did not increase survival from RML infection (Ahn et al, unpublished data). We will use BrnAggs to explore the behavior of multiple other scrapie prion strains and, in appropriate transgenic mice expressing human PrPC, test CreutzfeldtJakob disease prions. Our first goal will be to determine whether, unlike neurospheres, BrnAggs are susceptible to different prion strains. The ultimate goal is to use an in vitro system to test anti-prion drugs and gene therapies for human prion diseases. Brain aggregates will elucidate cellular and molecular pathobiology of prion diseases and other neurodegenerative disorders. Because it is possible to obtain meaningful biology and drug data in approximately 40 days with BrnAggs, this is more efficient than obtaining comparable data from mice that take 100 to 200 days to obtain. ACKNOWLEDGMENTS The authors thank Dr Stanley Prusiner for animals and PrP antibodies and Dr Lynn Pulliam for her help with learning the BrnAgg culture methods. REFERENCES 1. DeLong GR. Histogenesis of fetal mouse isocortex and hippocampus in reaggregating cell cultures. Dev Biol 1970;22:563Y83 2. Pulliam L, Dix RD, Panitch HS, et al. Use of aggregating brain cultures to study the replication of herpes simplex virus types 1 and 2 in central nervous system tissue. J Virol Methods 1984;9:301Y16 3. Pulliam L, Berens ME, Rosenblum ML. A normal human brain cell aggregate model for neurobiological studies. J Neurosci Res 1988;21:521Y30 4. Rempel HC, Pulliam L. HIV-1 Tat inhibits neprilysin and elevates amyloid beta. AIDS 2005;19:127Y35 5. Rempel H, Calosing C, Sun B, et al. Sialoadhesin expressed on IFNinduced monocytes binds HIV-1 and enhances infectivity. PLoS ONE 2008;3:e1967 6. Marshall GP 2nd, Reynolds BA, Laywell ED. Using the neurosphere assay to quantify neural stem cells in vivo. Curr Pharm Biotechnol 2007;8:141Y45 7. Marshall GP 2nd, Ross HH, Suslov O, et al. Production of neurospheres from CNS tissue. Methods Mol Biol 2008;438:135Y50 8. Giri RK, Young R, Pitstick R, et al. Prion infection of mouse neurospheres. Proc Natl Acad Sci USA 2006;103:3875Y80 9. Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science 1992; 255:1707Y10 10. Jensen JB, Parmar M. Strengths and limitations of the neurosphere culture system. Mol Neurobiol 2006;34:153Y61 11. Herva ME, Relano-Gines A, Villa A, Torres JM. Prion infection of differentiated neurospheres. J Neurosci Methods 2010;188:270Y75 12. Bosque PJ, Prusiner SB. Cultured cell sublines highly susceptible to prion infection. J Virol 2000;74:4377Y86 13. Enari M, Flechsig E, Weissmann C. Scrapie prion protein accumulation by scrapie-infected neuroblastoma cells abrogated by exposure to a prion protein antibody. Proc Natl Acad Sci USA 2001;98:9295Y99 14. Korth K, May BCH, Cohen FE, et al. Acridine and phenothiazine derivatives as pharmacotherapeutics for prion disease. Proc Natl Acad Sci USA 2001;98:9836Y41 15. Nakajima M, Yamada T, Kusuhara T, et al. Results of quinacrine administration to patients with Creutzfeldt-Jakob disease. Dement Geriatr Cogn Disord 2004;17:158Y63 16. Collins SJ, Lewis V, Brazier M, et al. Quinacrine does not prolong survival in a murine Creutzfeldt-Jakob disease model. Ann Neurol 2002;52: 503Y6 17. Spilman P, Lessard P, Sattavat M, et al. A F-secretase inhibitor and quinacrine reduce prions and prevent dendritic degeneration in murine brains. Proc Natl Acad Sci USA 2008;105:10595Y600 18. Collinge J, Gorham M, Hudson F, et al. Safety and efficacy of quinacrine in human prion disease (PRION-1 study): A patient-preference trial. Lancet Neurol 2009;8:334Y44 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. 465 Bajsarowicz et al J Neuropathol Exp Neurol Volume 71, Number 5, May 2012 19. Rubenstein R, Carp RI, Callahan SH. In vitro replication of scrapie agent in a neuronal model: Infection of PC12 cells. J Gen Virol 1984;65:2191Y98 20. Taraboulos A, Serban D, Prusiner SB. Scrapie prion proteins accumulate in the cytoplasm of persistently infected cultured cells. J Cell Biol 1990; 110:2117Y32 21. Schätzl HM, Laszlo L, Holtzman DM, et al. A hypothalamic neuronal cell line persistently infected with scrapie prions exhibits apoptosis. J Virol 1997;71:8821Y31 22. Cronier S, Laude H, Peyrin JM. Prions can infect primary cultured neurons and astrocytes and promote neuronal cell death. Proc Natl Acad Sci USA 2004;101:12271Y76 23. Archer F, Bachelin C, Andreoletti O, et al. Cultured peripheral neuroglial cells are highly permissive to sheep prion infection. J Virol 2004;78:482Y90 24. Ishikura N, Clever J, Bouzamondo-Bernstein E, et al. Notch-1 activation and dendritic atrophy in prion diseases. Proc Natl Acad Sci USA 2005; 102:886Y91 25. Klingenstein R, Lober S, Kujala P, et al. Tricyclic antidepressants, quinacrine and a novel, synthetic chimera thereof clear prions by destabilizing detergent-resistant membrane compartments. J Neurochem 2006;98:748Y59 26. Vaskovsky A, Lupowitz Z, Erlich S, et al. ErbB-4 activation promotes neurite outgrowth in PC12 cells. J Neurochem 2000;74:979Y87 27. Ni C-Y, Murphy MP, Golde TE, et al. Gamma-secretase cleavage and nuclear localization of ErbB-4 receptor tyrosine kinase. Science 2001; 295:2179Y81 28. Inoue E, Deguchi-Tawarada M, Togawa A, et al. Synaptic activity prompts gamma-secretase-mediated cleavage of EphA4 and dendritic spine formation. J Cell Biol 2009;185:551Y64 29. Heiseke A, Aguib Y, Schatzl HM. Autophagy, Prion Infection and their Mutual Interactions. Curr Issues Mol Biol 2009;12:87Y98 30. Ravikumar B, Berger Z, Vacher C, et al. Rapamycin pre-treatment protects against apoptosis. Hum Mol Genet 2006;15:1209Y16 31. Carloni S, Buonocore G, Balduini W. Protective role of autophagy in neonatal hypoxia-ischemia induced brain injury. Neurobiol Dis 2008;32:329Y39 32. Ravikumar B, Duden R, Rubinsztein DC. Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 2002;11:1107Y17 33. Webb JL, Ravikumar B, Atkins J, et al. Alpha-synuclein is degraded by both autophagy and the proteasome. J Biol Chem 2003;278:25009Y13 34. Williams A, Jahreiss L, Sarkar S, et al. Aggregate-prone proteins are cleared from the cytosol by autophagy: Therapeutic implications. Curr Top Dev Biol 2006;76:89Y101 35. Berger Z, Ravikumar B, Menzies FM, et al. Rapamycin alleviates toxicity of different aggregate-prone proteins. Hum Mol Genet 2006;15:433Y42 36. Fornai F, Longone P, Ferrucci M, et al. Autophagy and amyotrophic lateral sclerosis: The multiple roles of lithium. Autophagy 2008;4:527Y30 37. Imaida K, Yoshida J, Uneyama C, et al. Dose-dependent enhancing effects of quinacrine on induction of preneoplastic glutathione S-transferase placental form positive liver cell foci in male F344 rats. Carcinogenesis 1991;12:1911Y15 38. Gupta A, Roy S, Lazar AJ, et al. Autophagy inhibition and antimalarials promote cell death in gastrointestinal stromal tumor (GIST). Proc Natl Acad Sci U S A 2010;107:14333Y38 39. Jendroska K, Heinzel FP, Torchia M, et al. Proteinase-resistant prion protein accumulation in Syrian hamster brain correlates with regional pathology and scrapie infectivity. Neurology 1991;41:1482Y90 40. Tatzelt J, Groth DF, Torchia M, et al. Kinetics of prion protein accumulation in the CNS of mice with experimental scrapie. J Neuropathol Exp Neurol 1999;58:1244Y49 41. Safar J, Wille H, Itri V, et al. Eight prion strains have PrPSc molecules with different conformations. Nature Med 1998;4:1157Y65 42. Safar JG, Lessard P, Tamguney G, et al. Transmission and detection of prions in feces. J Infect Dis 2008;198:81Y89 43. DeArmond SJ, Bajsarowicz K. PrPSc accumulation in neuronal plasma membranes links Notch-1 activation to dendritic degeneration in prion diseases. Mol Neurodegener 2010;5:6 44. Suslov ON, Kukekov VG, Ignatova TN, et al. Neural stem cell heterogeneity demonstrated by molecular phenotyping of clonal neurospheres. Proc Natl Acad Sci USA 2002;99:14506Y11 45. Drake KR, Kang M, Kenworthy AK. Nucleocytoplasmic distribution and dynamics of the autophagosome marker EGFP-LC3. PLoS One 2010;5:e9806 46. Mann SS, Hammarback JA. Molecular characterization of light-chain 3. A microtubule binding subunit of MAP1A and MAP1B. J Biol Chem 1994;269:11492Y97 47. Mann SS, Hammarback JA. Gene localization and developmental expression of light-chain 3: A common subunit of microtubule-associated protein 1A(MAP1A) and MAP1B. J Neurosci Res 1996;43:535Y44 48. Seidenbecher CI, Landwehr M, Smalla KH, et al. Caldendrin but not calmodulin binds to light-chain 3 of MAP1A/B: An association with the microtubule cytoskeleton highlighting exclusive binding partners for neuronal Ca(2+)-sensor proteins. J Mol Biol 2004;336:957Y70 49. Bouzamondo-Bernstein E, Hopkins SD, Spillman P, et al. The neurodegenerative sequence in prion diseases: Evidence from functional, morphological and ultrastructural studies of the GABAergic system. J Neuropathol Exp Neurol 2004;63:882Y99 50. Caughey B. Prion protein interconversions. Philos Trans R Soc Lond B Biol Sci 2001;356:197Y200, Discussion 202 51. Lampert P, Hooks J, Gibbs CJ Jr, et al. Altered plasma membranes in experimental scrapie. Acta Neuropathol (Berl) 1971;19:81Y93 52. Beck E, Bak IJ, Christ JF, et al. Experimental kuru in the spider monkey. Histopathological and ultrastructural studies of the brain during early stages of incubation. Brain 1975;98:595Y612 53. Jeffrey M, Halliday WG, Bell J, et al. Synapse loss associated with abnormal PrP precedes neuronal degeneration in the scrapie-infected hippocampus. Neuropath Appl Neurobiol 2000;26:41Y54 54. Bouzamondo-Bernstein E, Milroy AM, Ralston HJ III, et al. Selective neuronal vulnerability of GABAergic neurons during experimental scrapie infection: Insights from an ultrastructural investigation. Brain Res 2000; 874:210Y15 55. Sikorska B, Liberski PP, Giraud P, et al. Autophagy is a part of ultrastructural synaptic pathology in Creutzfeldt-Jakob disease: A brain biopsy study. Int J Biochem Cell Biol 2004;36:2563Y73 56. Heiseke A, Aguib Y, Riemer C, et al. Lithium induces clearance of protease resistant prion protein in prion-infected cells by induction of autophagy. J Neurochem 2009;109:25Y34 57. Ghaemmaghami S, Ahn M, Lessard P, et al. Continuous quinacrine treatment results in the formation of drug-resistant prions. PLoS Pathog 2009;5:e1000673 58. Iso T, Kedes L, Hamamori Y. HES and HERP families: Multiple effectors of the Notch signaling pathway. J Cell Physiol 2003;194:237Y55 59. Taraboulos A, Raeber AJ, Borchelt DR, et al. Synthesis and trafficking of prion proteins in cultured cells. Mol Biol Cell 1992;3:851Y63 60. Liberski PP. Spongiform changeYan electron microscopic view. Folia Neuropathol 2004;42(Suppl B):59Y70 61. Zeng M, Zhou JN. Roles of autophagy and mTOR signaling in neuronal differentiation of mouse neuroblastoma cells. Cell Signal 2008;20: 659Y65 62. Pfeffer S. Vesicle tethering factors united. Mol Cell 2001;8:729Y30 63. Yu WH, Cuervo AM, Kumar A, et al. MacroautophagyVA novel Betaamyloid peptide-generating pathway activated in Alzheimer’s disease. J Cell Biol 2005;171:87Y98 64. Nixon RA, Wegiel J, Kumar A, et al. Extensive involvement of autophagy in Alzheimer disease: An immuno-electron microscopy study. J Neuropathol Exp Neurol 2005;64:113Y22 65. Fevrier B, Vilette D, Archer F, et al. Cells release prions in association with exosomes. Proc Natl Acad Sci USA 2004;101:9683Y88 66. Porto-Carreiro I, Fevrier B, Paquet S, et al. Prions and exosomes: From PrPc trafficking to PrPsc propagation. Blood Cells Mol Dis 2005;35:143Y48 67. de Gassart A, Geminard C, Fevrier B, et al. Lipid raft-associated protein sorting in exosomes. Blood 2003;102:4336Y44 68. Pfeffer SR. Unconventional secretion by autophagosome exocytosis. J Cell Biol 2010;188:451Y42 69. Duran JM, Anjard C, Stefan C, et al. Unconventional secretion of Acb1 is mediated by autophagosomes. J Cell Biol 2010;188:527Y36 70. Nickel W, Rabouille C. Mechanisms of regulated unconventional protein secretion. Nature Rev 2009;10:148Y55 71. Giese A, Brown DR, Groschup MH, et al. Role of microglia in neuronal cell death in prion disease. Brain Pathol 1998;8:449Y57 72. Dello Russo C, Lisi L, Tringali G, et al. Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem Pharmacol 2009;78:1242Y51 466 Ó 2012 American Association of Neuropathologists, Inc. Copyright © 2012 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.