Skip to main content
Erschienen in: Molecular Neurodegeneration 1/2018

Open Access 01.12.2018 | Research article

Structural and mechanistic aspects influencing the ADAM10-mediated shedding of the prion protein

verfasst von: Luise Linsenmeier, Behnam Mohammadi, Sebastian Wetzel, Berta Puig, Walker S. Jackson, Alexander Hartmann, Keiji Uchiyama, Suehiro Sakaguchi, Kristina Endres, Jörg Tatzelt, Paul Saftig, Markus Glatzel, Hermann C. Altmeppen

Erschienen in: Molecular Neurodegeneration | Ausgabe 1/2018

Abstract

Background

Proteolytic processing of the prion protein (PrPC) by endogenous proteases generates bioactive membrane-bound and soluble fragments which may help to explain the pleiotropic roles of this protein in the nervous system and in brain diseases. Shedding of almost full-length PrPC into the extracellular space by the metalloprotease ADAM10 is of peculiar relevance since soluble PrP stimulates axonal outgrowth and is protective in neurodegenerative conditions such as Alzheimer’s and prion disease. However, molecular determinates and mechanisms regulating the shedding of PrP are entirely unknown.

Methods

We produced an antibody recognizing the neo-epitope of shed PrP generated by ADAM10 in biological samples and used it to study structural and mechanistic aspects affecting the shedding. For this, we investigated genetically modified cellular and murine models by biochemical and morphological approaches.

Results

We show that the novel antibody specifically detects shed PrP in cell culture supernatants and murine brain. We demonstrate that ADAM10 is the exclusive sheddase of PrPC in the nervous system and reveal that the glycosylation state and type of membrane-anchorage of PrPC severely affect its shedding. Furthermore, we provide evidence that PrP shedding can be modulated by pharmacological inhibition and stimulation and present data suggesting that shedding is a relevant part of a compensatory network ensuring PrPC homeostasis of the cell.

Conclusions

With the new antibody, our study introduces a new tool to reliably investigate PrP-shedding. In addition, this study provides novel and important insight into the regulation of this cleavage event, which is likely to be relevant for diagnostic and therapeutic approaches even beyond neurodegeneration.
Begleitmaterial
Additional file 2: (.jpg) Species specificity of the sPrPG228 antibody. (A) Comparison of the C-terminal amino acid sequence of PrPC in mouse (Mus musculus), rat (Rattus norvegicus) and rabbit (Oryctolagus cuniculus) (source: www.​uniprot.​org). “NH 2 -…-” indicates the N-terminal direction, “-GPI” the C-terminal GPI-anchor attachment site. Asterisks indicate position of ADAM10-mediated shedding in mice and rats with Gly228 representing the new C-terminus of shed PrP. Note the sequence difference compared with rabbit PrPC. (B) Western blot analysis of forebrain homogenates from different murine models (tga20, Prnp0/0, wild-type (C57BL/6)) as well as from rat and rabbit. As expected for its epitope, the sPrPG228 antibody detects sPrP in mouse (tga20 and wild-type) and rat, whereas the brain sample of rabbit only presents an immunoglobulin light chain (rb Ig-LC) signal at 25 kDa resulting from the anti-rabbit secondary antibody used for detection. Re-probing the blot with POM2 antibody reveals expression levels of PrPC. (JPEG 388 kb)
Additional file 6: (.jpg) Side-by-side comparison of SWA and TM treatments and enzymatic deglycosylation reactions. Western blot of untreated (untr.), SWA- or TM-treated N2a cells showing lysates without (−) or with (+) enzymatic treatment for differential deglycosylation (Endo H or PNGase F). As also shown in Fig. 3a and b, TM-treatment causes a complete inhibition of PrP glycosylation, whereas SWA-treatment results in a shift in the banding pattern (compared to untreated cells) and (at least partial) Endo H sensitivity due to inhibition of complex glycosylation. Changes in the glycopattern and running behaviour support the functioning of our enzymatic deglycosylation protocols also performed for the experiments shown in Figs. 2, 3 and 4. Actin is shown as a loading control. (JPEG 221 kb)
Additional file 9: (.jpg) Quantification of fl-PrP levels and ratio of sPrP/fl-PrP in Sort1 knockout mice. These quantifications refer to main Fig. 5j. (A) Increased amounts of fl-PrP are found in brains of Sort1 KO mice (2.11 ± 0.23; p = 0.0004; n = 4) compared to controls (WT set to 1.00 ± 0.21; SD). Actin served as loading control and for reference in densitometric quantification. (B) No significant differences are detected in the ratio of sPrP to fl-PrP between Sort1 KO mouse brains (0.85 ± 0.07, p = 0.128; n = 4) and controls (WT set to 1.00 ± 0.14). (JPEG 229 kb)
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13024-018-0248-6) contains supplementary material, which is available to authorized users.
Abkürzungen
Ab
antibody
ADAM
a disintegrin and metalloproteinase
AIDS
acquired immunodeficiency syndrome
APP
Amyloid precursor protein
Amyloid-beta peptide
BSA
bovine serum albumin
BSE
bovine spongiform encephalopathy
cKO
conditional knockout
d.n.
dominant negative
DMSO
dimethyl sulfoxide
FBS
fetal bovine serum
fl-PrP
full length prion protein
GI
GI254023X (ADAM10 inhibitor)
GPI
glycosylphosphatidylinositol
HIV
human immunodeficiency virus
KI
knock-in
KO
knockout
MDCK
Madin-Darby Canine Kidney (cells)
MEF
murine embryonal fibroblasts
NaDOC
sodium deoxycholate
PBS
phosphate buffered saline
PI
protease inhibitor
PI-PLC
phosphatidylinositol-specific phospholipase C
Prnp 0/0
PrPC knockout
PrPC
cellular prion protein
PrPSc
pathological (Scrapie) isoform of the prion protein
PrP-TM
transmembrane PrP
Sort1
sortilin-1
sPrP
shed PrP
sPrPG228
antibody against shed PrP
SWA
swainsonine
TACE
tumor necrosis factor α converting enzyme (ADAM17)
TALEN
Transcription Activator-like Effector Nuclease
TBS
Tris-buffered saline
TCA
trichloroacetic acid
tga20
prion protein overexpressing mouse line
TM
Tunicamycin
WT
wild-type

Background

Proteolytic processing is an essential regulator of protein function and differs from many other posttranslational modifications by its irreversible character. As exemplified decades ago in the case of prohormones (such as the proopiomelanocortin [1]), differential or subsequent cleavages by endogenous proteases produce fragments with intrinsic biological functions, differing from the ones of the larger precursors. This concept may also help in understanding and explaining the biology of other “multifunctional” proteins, i.e. proteins with more than just one particular function ascribed to them.
One of these proteins is the cellular prion protein (PrPC), for which a multitude of physiological functions has been suggested in different tissues, cells and experimental settings [2, 3], even though not in each case without controversy or questionable reproducibility [4, 5]. For instance, PrPC has been linked to developmental processes [6, 7], cell adhesion [8, 9], neurite outgrowth, axon guidance and synapse formation [1014], as well as to neuroprotection [1517] and regulation of the circadian rhythm [18]. Among the currently best characterized functions are its contributions to myelin maintenance [4, 1921] and cellular homeostasis of divalent ions [22, 23] as well as its involvement in signaling events [2426].
Too many functional implications for just one protein? Not necessarily. While transient interactions of PrPC with alternating binding partners in different cellular locations may partially account for this functional diversity [5, 27], so might its proteolytic processing [28]. In fact, different highly conserved cleavage events occur constitutively on a relevant fraction of PrPC [2931], yet scientists are just starting to understand their biological relevance.
In contrast to some of the suggested physiological functions, the relevance of PrPC in neurodegenerative proteinopathies is widely accepted. First and foremost, it is the essential substrate for the process of templated misfolding underlying fatal and transmissible prion diseases, such as Creutzfeldt-Jakob disease in humans or BSE in cattle [3234]. Once having adopted its pathogenic conformation (PrPSc), the prion protein is the key component of the infectious particles termed prions [32, 3537]. Second, binding of toxic oligomeric protein species, such as PrPSc (in prion diseases [38]), Aβ (in Alzheimer’s disease [3942]) or α-synuclein (in Parkinson’s disease [43, 44]), to PrPC at the neuronal surface results in neurotoxic signaling. As for the physiological functions, increasing evidence suggests that proteolytic cleavages also impact on these pathogenic roles of the prion protein [28, 45, 46].
Here, we focus on the most membrane-proximate cleavage of PrPC, i.e. its shedding from the neuronal surface and release into the extracellular space by the metalloprotease ADAM10 [47, 48]. This cleavage not only regulates membrane levels of PrPC and, thus, PrPC-related functions at the neuronal surface [28]. The resulting soluble fragment, shed PrP, likely has intrinsic functions as supported by studies using (recombinant) anchorless analogues, that showed beneficial effects with regard to axon outgrowth and synapse formation [13, 14] or neuroprotection [15, 49]. Focusing on neurodegeneration, we have recently shown a significant impact on the course of prion disease in mice by conditional depletion of the sheddase ADAM10 [50, 51], as have others by overexpression of exogenous ADAM10 [52] or by transgenic expression of anchorless versions of PrP [53, 54]. Moreover, by reducing membrane-bound PrPC as a receptor and by producing anchorless PrP, which can block and detoxify Aβ and other harmful protein species in the extracellular space [5558], shedding may also have a protective role in other, more frequent proteinopathies [45].
Surprisingly, shed PrP has recently been associated with the development of specific tumours in the nervous system, where it correlates with increased cancer cell proliferation [59]. In addition, a recent report shows critical involvement of shed PrP in the neuropathogenesis of HIV/AIDS by recruiting monocytes and aggravating the inflammatory response and the associated cognitive impairment [60].
Thus, given that shedding of PrPC might provide a promising and potent target for therapy of various pathological conditions, a deeper mechanistic understanding and knowledge of factors influencing this cleavage is required. Here, we first introduce and characterize a novel antibody detecting shed PrP with high specificity and sensitivity. Using this tool, we investigate different structural (i.e. glycosylation state and membrane anchorage) and mechanistic aspects in vitro and in vivo for how they impact on this relevant proteolytic event. Finally, we show that shedding is part of a compensatory cellular network regulating PrPC homeostasis.

Methods

Plasmids

The following constructs were used for transient transfection of cells. Detailed descriptions of the constructs can be found in the corresponding references: PrP-WT, PrPC glycomutants PrP-G1, PrP-G2, PrP-G3 and anchor-mutant PrPGPI-Thy1 [61], PrP-TM (PrP-CD4 [62, 63]). All PrP constructs contained the 3F4 tag [64]. The N-terminally truncated PrP-C1 construct was cloned from the plasmid pcDNA3.1(+)/Zeo containing the murine Prnp gene. The sequence coding for the N-terminal part of PrPC (aa23–110) was deleted by use of the restriction enzymes XbaI and HindIII and the resulting construct (Δaa23–110; i.e. PrP-C1) was verified by DNA sequencing.

Antibody production

The sPrPG228 antibody for the specific detection of shed PrP was generated by use of an anti-peptide approach and the classical 87-day polyclonal protocol (Eurogentec, Belgium). Briefly, based on the sequence information of murine PrPC and previous determination of the cleavage site for ADAM10 [47], a recombinant peptide NH2-C-QAYYDG-COOH (in which G-COOH represents G228 as the new C-terminus of shed PrP exposed after cleavage (Fig. 1a)) was produced and N-terminally coupled to Megathura crenulata keyhole limpet hemocyanin (KLH) as carrier protein. This peptide was used as immunogen and injected into rabbit at days 0, 14, 28 and 56 of the programme. Bleedings were done at days 0, 38 and 66 to investigate the success of the immunization process by standardized ELISA tests. Animals were sacrificed and final bleeds were obtained at day 87. Standardized quality measures and affinity purification were performed at Eurogentec. Importantly, a second peptide (NH2-C-KESQAYYDGRRS-COOH) mimicking the C-terminus of fl-PrP (without the GPI anchor) was produced, coupled to a resin and served as a “negative control” to eliminate all antibodies from the polyclonal serum that would otherwise bind to fl-PrP.

Rodent brain samples

Use of animal material in this study was in strict compliance with the Guide for the Care and Use of Laboratory Animals and ethics guidelines of the responsible local authorities. Frozen forebrain samples from wild-type C57BL/6, prion protein deficient (Prnp0/0 [65]), prion protein overexpressing (tga20 [66]) mice as well as from mice with conditional knockout of ADAM10 in forebrain neurons (A10 cKO and wild-type littermate controls [67]), with transgenic overexpression of dominant negative ADAM10 (A10 d.n. and wild-type controls [68]), with depletion of sortilin-1 (Sort1 KO and wild-type controls [69]) or with a knock-in of 3F4-tagged PrPC (PrP3F4KI and controls; both had a 192S4 background [70]), and from a rat and a rabbit were used to prepare 10% (w/v) homogenates in RIPA buffer (50 mM Tris-HCl pH 8, 150 mM NaCl, 1% NP-40, 0.5% Na-Deoxycholate, 0.1% SDS) freshly supplemented with Complete EDTA-free protease (PI) and PhosStop phosphatase inhibitor cocktails (Roche) on ice. Samples were homogenized with 30 strokes using a dounce homogenizer and incubated on ice for 20 min, shortly vortexed and incubated for another 20 min before centrifugation at 12,000 g at 4 °C for 12 min. Total protein content was assessed by Bradford assay (BioRad). Supernatants were either further processed for SDS-PAGE or stored at − 80 °C.

Cell culture, transfection and treatments

Murine neuroblastoma cells (N2a) and mouse embryonic fibroblasts (MEF; [71]) were maintained at 37 °C under an atmosphere of 5% CO2 in Dulbecco’s modified Eagle’s medium (DMEM; Thermo Fisher Scientific) supplemented with 10% fetal bovine serum (FBS; Thermo Scientific Fisher). N2a PrP-KO cells were generated using the TALEN approach and characterized in detail before [72]. N2a PrP-KO cells were transfected using Lipofectamine 2000 (Thermo Fisher Scientific) following the manufacturer’s instructions. For stable overexpression of PrP3F4 in N2a PrP-KO (used for the glycopattern analysis shown in Fig. 1h) cells were kept for 3 weeks in selection media (Zeocin 400 μg/ml; Thermo Fisher Scientific) and single resistant clones were selected for amplification.
Treatments of cells were performed by adding the following compounds (and concentrations) to the cell culture media: Resveratrol (20 μM), Tamibarotene/Am80 (1 μM), GI254023X (3 μM), Tunicamycin (2.5 μg/ml), Swainsonine (5 μg/ml), Leupeptin (200 μg/ml). All compounds were purchased from Merck. These treatments were carried out in 6-well plates with 1 ml OptiMEM for 18 h overnight. In the case of Tunicamycin and Swainsonine cells were pretreated for 8 h. Treatment with GM6001 (25 μM) or Batimastat (10 μM) was for 10 h.

Treatment of cells with PI-PLC

Two days post-transfection cells (grown in 6-well plate format) were incubated with 0.5 U/ml Phospholipase C (PI-PLC; Sigma-Aldrich) in 1 ml OptiMEM for 2 h at 37 °C, 5% CO2 in order to cleave GPI-anchor structures and release GPI-anchored proteins from the cellular surface. Supernatants were subsequently harvested and further processed while cells were lysed as described below.

PNGase F and Endo H digestion

For removal as well as for investigations on processing and maturation of N-linked glycans attached to PrPC, cell lysates and/or supernatants were digested with either PNGase F or Endo H (New England Biolabs) according to the manufacturer’s protocols.

Sample preparation, TCA precipitation, cell surface biotinylation assay, SDS-PAGE and western blot analysis

N2a cells were washed with PBS and lysed with RIPA buffer, incubated on ice for 15 min before centrifugation at 12,000 g for 12 min at 4 °C. The protein content of the resulting supernatant was determined by Bradford assay. Prior to SDS-PAGE, cell lysates or brain homogenates (see above) were mixed with 4× loading buffer (including β-mercaptoethanol) and denatured for 6 min at 96 °C.
For the analysis of cell culture supernatants, experiments were carried out with serum-free media (OptiMEM). Supernatants were precipitated with trichloroacetic acid (TCA). For this, supernatants were collected and immediately incubated with already dissolved protease inhibitor cocktail, cleared from dead cells and debris by mild centrifugations at 500 g and 5.000 g for 5 min each. 1/100 volume of 2% sodium deoxycholate (NaDOC) was then added and each sample was shortly vortexed. After 30 min incubation on ice, samples were mixed with 1/10 volume of 100% TCA and again incubated for 30 min on ice. After centrifugation at 15,000 g for 15 min at 4 °C, the supernatant was aspirated, and the air-dried pellet was dissolved in 1× loading buffer and boiled for 6 min at 96 °C.
For labelling and purification of proteins at the cell surface, a surface biotinylation assay was performed as described earlier [50] prior to cell lysis.
For SDS-PAGE, denatured samples were loaded on either precast Nu-PAGE 4–12% Bis-Tris protein gels (Thermo Fisher Scientific) or self-made 10% or 12% SDS-gels. After electrophoretic separation, proteins were transferred to nitrocellulose membranes (BioRad) by wet-blotting and membranes were subsequently blocked for 1 h with 5% skimmed dry milk dissolved in TBS-T (containing 0.1% Tween-20) and incubated with primary antibody diluted in 5% skimmed dry milk in TBS-T overnight at 4 °C on a shaking platform. For detection of full length PrPC (fl-PrP), mouse monoclonal antibodies POM1 (1 μg/ml), POM2 (0.6 μg/ml) [73] or, in the case of the sortilin-1 knockout mouse brains (Fig. 5j), SAF61 (0.2 μg/ml; Bertin Pharma) were used. Proteolytically shed PrPC was detected with our new rabbit polyclonal sPrPG228 antibody (0.2 μg/ml) characterized in detail herein. Moreover, we used anti-ADAM10 (0.4 μg/ml; abcam), anti-mouse β-amyloid antibody for detection of sAPPα (1 μg/ml; BioLegend), anti-actin antibody clone C4 (MAB1501, 1:1000; Merck) and anti-Flotillin-1 clone 18 (0.25 μg/ml; BD Biosciences). Membranes were subsequently washed with TBS-T and incubated for 1 h with respective HRP-conjugated secondary antibodies and subsequently washed 6× with TBS-T. After incubation with Pierce ECL Pico or Super Signal West Femto substrate (Thermo Fisher Scientific), chemiluminescence was detected with a ChemiDoc imaging station (BioRad) and densitometrically quantified using Image Studio Lite software version 5.2 (LI-COR).

Immunofluorescence staining of surface proteins and microscopy

N2a cells were grown on glass coverslips. After washing with PBS, living cells were incubated for 20 min on ice (to avoid endocytosis) with the primary antibody dissolved in 2% BSA/PBS. Surface PrPC was detected with POM1 antibody (10 μg/ml). After several washes with PBS, cells were incubated with suitable secondary antibodies for 20 min on ice, subsequently fixed in 4% paraformaldehyde for 20 min at room temperature and mounted on glass slides with DAPI Fluoromount G (Southern Biotech). Analysis was performed using a TCS SP5 confocal microscope (Leica).

Histological and immunohistochemical stainings

Sampling, formalin fixation, paraffin embedding, hematoxilin and eosin (H&E) staining as well as immunostaining with anti-prion protein antibody SAF84 (Caiman Chemical) of murine brain samples has been described earlier [50]. Immunostaining of shed PrP was likewise performed in one run using a Benchmark XT machine (Ventana) to allow for best comparability. In brief, deparaffinated brain sections were boiled for 1 h in citrate buffer (CC1 Cell Conditioning Solution, Ventana) for antigen retrieval and then incubated for 1 h with the sPrPG228 primary antibody (7 μg/ml; in antibody diluent solution (Zytomed) with 5% goat serum). Detection with anti-rabbit secondary antibody (Nichirei Biosciences) and Ultra View Universal DAB Detection kit (Ventana), as well as blue counterstaining were performed by the machine following standardized protocols.

Exosome isolation and nanoparticle tracking analysis

N2a cells were cultured in OptiMEM for 18 h. For the harvest of extracellular vesicles (here further referred to as exosomes), cell culture supernatants were first complemented with PI and centrifuged for 10 min at 1000 g and further at 7500 g for 15 min at 4 °C, followed by filtration through a 22 μm membrane to clear it from dead cells and debris. Exosomes were then pelleted by ultracentrifugation at 100,000 g for 70 min at 4 °C in an Optima L-100 XP using a SW40Ti rotor (Beckman Coulter, Inc.) and subsequently resuspended in PBS containing PI. For quantification and characterization, a NanoSight LM14 (Malvern Instruments) equipped with a 638 nm laser and a Marlin F033B IRF camera (Allied Vision Technologies) was used. For each sample, 10 videos of 10 s length were recorded with a camera intensity setting of 16 and analysed to assess average size and concentration of exosomes using the batch processing function of the software. For normalized western blot analysis, 5 × 1010 exosomes per sample were used.

Statistical analysis

For experiments using mouse brain samples, n refers to the number of biological samples (i.e. mice) per experimental group. For cell culture-based data, n stands for the number of independent experiments. Statistical comparison of western blot quantifications was performed using Student’s t-test and significance was considered with p-values as follows: *p < 0.05, **p < 0.005, ***p < 0.001.

Results

A novel antibody specifically detects shed PrP and reveals important insight into the ADAM10-mediated shedding of PrPC in mice and cells

ADAM10 is the relevant sheddase of PrPC releasing a soluble form (shed PrP, sPrP) from the plasma membrane [47, 48]. Since membrane-bound full length (fl) PrPC and its shed form only differ in three amino acids (murine sequence) and the GPI-anchor, it is hard to reliably discriminate between both in most approaches. Based on available sequence information and the previous identification of the cleavage site for ADAM10 [47], we therefore generated an antibody specific for sPrP (sPrPG228) being directed against the newly generated carboxy-terminus at Glycine 228 (G228) exposed after cleavage (Fig. 1a).
To characterize this antibody in detail, we analyzed forebrain homogenates of age-matched Prnp0/0 mice (as negative control), recently described mice with neuron-specific (CamKIIα-driven) depletion of ADAM10 (A10 cKO; to control for specificity) as well as wild-type littermate controls, and PrPC-overexpressing tga20 mice (as positive controls) by western blot. As expected, detection with our new antibody consistently revealed no signal in Prnp0/0 samples, basal levels in wild-type mice and strongly increased signal intensity in tga20 mice (Fig. 1b). Though we expected significantly reduced levels of shed PrP in A10 cKO mice, to our surprise we could not detect any signal in these samples. Besides supporting the specificity of the antibody, this indicates that no other cell types or proteases contribute to this cleavage in brain. Re-probing the same blot with an antibody against fl-PrP revealed an increase in total PrPC levels in A10 cKO mouse brains (Fig. 1b), a finding that has been made earlier and can be attributed to the lack of shedding [50]. Moreover, while this blotting strategy demonstrated the overlapping banding pattern (as well as the masking of sPrP signals by excess amounts of fl-PrP using common PrP antibodies), it also revealed a small shift in the molecular weight of sPrP corresponding to the lack of three amino acids and the GPI-anchor (Fig. 1b and Additional file 1). We also investigated the species specificity of the new antibody using mouse, rat and rabbit brain samples. As expected for the different C-terminal PrPC sequences, the sPrPG228 antibody only detected sPrP in brain homogenates of mice and rat (Additional file 2).
Though not being in the focus of this study, we were also interested in the applicability of the antibody in morphological analyses and performed immunohistochemical staining of paraffin-embedded mouse brain sections. As exemplified for the hippocampal area in Additional file 3, no signal was obtained in a Prnp0/0 mouse, whereas a diffuse staining was found in wild-type and, with higher intensity, in tga20 brain as could be expected for a soluble fragment distributed in the brain parenchyma.
Although, structurally, all three glycoforms of PrPC can be shed (as demonstrated in tga20 brain (Fig. 1c)), a strong predominance of the diglycosylated form of sPrP was obvious in all of our biochemical analyses. To investigate this in more detail, we analyzed the glycopattern of sPrP compared to cell-associated fl-PrP in brain homogenates of wild-type mice (Fig. 1d) and found a clear shift and a drastic increase in the proportion of diglycosylated sPrP (mean: 97 ± 1%; compared to 60 ± 4% for fl-PrP; n = 3; ±SD) with only little mono- (3 ± 1%; fl-PrP: 33 ± 3%) and almost no unglycosylated sPrP (0.07 ± 0.03%; fl-PrP: 6.8 ± 0.8%).
As a model for downregulation of ADAM10-mediated cleavage events, we investigated PrP shedding in forebrains of mice overexpressing a dominant negative form of ADAM10 (A10 d.n.) in addition to the endogenous protease [68] (Fig. 1e). When referring the sPrP to the respective fl-PrP signal, we found a ~ 50% reduction (mean sPrP/fl-PrP ratio: 0.51 ± 0.05; n = 4; ±SEM) in A10 d.n. mice compared to matched controls (set to 1.00 ± 0.13).
Since for main parts of this study we used N2a cells transfected with murine versions of PrPC containing the 3F4 tag in the middle of the protein sequence, we first had to show that this modification does not influence the shedding event. This is even more important as it is known, that the course of prion diseases is altered by this tag [70, 74]. We therefore decided to investigate shedding in the best possible model, i.e. in PrP3F4 knock-in (KI) mice expressing levels of 3F4-tagged PrP identical to PrPC levels in wild-type mice (Additional file 1) [70]. No significant differences in sPrP levels were observed between controls (set to 1.00 ± 0.23; n = 3; ±SD) and PrP3F4 KI mice (0.85 ± 0.12) thus ruling out an impact of this modification on PrP shedding as could be expected from its intramolecular distance to the membrane-proximate shedding site.
We next employed the new antibody in cell culture-based experiments. Given that manipulation of PrPC shedding may become a therapeutic option in different pathologies, we investigated how pharmacological stimulation and inhibition of ADAM10 affect sPrP production in N2a cells (Fig. 1f,g). Among others, the stilbenoid resveratrol and the synthetic retinoid tamibarotene (Am80) have been successfully used to increase ADAM10-mediated cleavage events [75, 76]. We also found elevated levels of sPrP in supernatants of N2a cells treated with these substances compared to solvent-treated controls (Fig. 1f). In contrast, shedding was abolished upon treatment with the ADAM10-selective inhibitor GI254023X (GI) [77]. Of note, upon re-probing the “supernatant blot” with another PrP antibody (POM2), a strong signal was obtained under GI-treatment indicative of a release of fl-PrP by alternative routes when shedding is blocked (as discussed later). Fittingly, cell-associated PrPC levels (in lysates) remained rather unaffected by the different treatments further supporting existence of compensatory mechanisms regulating PrPC homeostasis in N2a cells (discussed later). We also assessed the metalloprotease inhibitors GM6001 and batimastat (Fig. 1g). These drugs likewise abolished the shedding of PrPC at the cell surface yet did not significantly alter production of N1 and C1 fragments resulting from the α-cleavage of PrPC. There is controversy regarding the involvement of ADAMs in the α-cleavage (reviewed in [45, 78]). However, due to the lack of membrane permeability of the inhibitors used here, this finding cannot count as an argument against ADAMs as potential “α-PrPases”, given that α-cleavage is thought to occur mainly within the secretory pathway [79]. Again, levels of cell-associated fl-PrP did not appear to be altered by these treatments.
Lastly, consistent with our findings in mouse brain (Fig. 1d), we also observed a changed glycopattern of sPrP compared to fl-PrP in N2a cells (Fig. 1h; diglycosylated: 84.2 ± 4.4% (sPrP) vs. 67.4 ± 0.9% (fl-PrP); monoglycosylated: 15.6 ± 4.2% (sPrP) vs. 29.6 ± 1.0% (fl-PrP); unglycosylated: 0.22 ± 0.18% (sPrP) vs. 2.9 ± 0.3% (fl-PrP); n = 3; ±SD) though relatively more monoglycosylated sPrP is found in N2a cells (15.6 ± 4.2%) than in brain (3 ± 1%; Fig. 1d). To clarify whether our findings indicate a real preference for the shedding of diglycosylated PrP or rather reflect the availability of different glycoforms at the plasma membrane, we performed cell surface biotinylation and glycopattern analysis in N2a cells (Additional file 4). Though relatively more diglycosylated PrP is indeed available at the plasma membrane (compared to total PrP levels in cell lysates; Additional file 4B), our data still argues in favor of a preference for diglycosylated PrP given the strong predominance of this form among shed PrP (Fig. 1d, h). In summary, we have generated a sensitive and highly specific antibody to discriminate between shed and fl-PrP in mouse brains and cell culture supernatants. ADAM10 on neurons seems to be the dominant (if not exclusive) PrP sheddase. ADAM10-mediated shedding of PrPC can be modulated by various means, and our shedding-specific antibody is a useful read-out tool for such experiments. Though all glycoforms can in principle be shed, diglycosylated PrP by far represents the major substrate for ADAM10.

The glycosylation state impacts on PrP shedding

Glycosylation of PrPC impacts on its biology and role in prion disease [62, 8083]. Given the predominance of diglycosylated sPrP under normal conditions (Fig. 1), we wondered how shedding would be affected if only specific glycoforms of PrPC are present in cells. To this end, we transfected PrPC-depleted N2a cells (PrP-KO; generated using the TALEN strategy and described earlier [72]) with either wild-type PrP or PrP glycomutants carrying a mutation in either the first (N180Q mutant; PrP-G1), the second (N196Q mutant; PrP-G2) or both (N180Q/N196Q mutant; PrP-G3) N-glycosylation sites and, thus, giving rise to mono- (G1 and G2) or unglycosylated (G3) PrPC. Using these glycomutants, we could previously demonstrate a relevant impact of the N-glycans on the sorting of PrPC in polarized cells [61]. Despite differences in transfection efficiencies, western blot analysis revealed the typical banding pattern for the glycomutants as observed for similar mutants in transgenic mice (Fig. 2a) [83]. Like fl-PrP, the membrane-attached C1 fragment resulting from α-cleavage of PrPC also presents with a three-banding pattern with the diglycosylated C1 overlapping with unglycosylated fl-PrP. For a better characterization, deglycosylation was performed to only obtain unglycosylated fl-PrP and C1 fragment and showed that all mutants undergo α-cleavage (Fig. 2b). No effect was observed upon treatment of lysates with Endo H indicating a correct processing of the glycans and trafficking out of the ER and to the cellular surface for all transfected mutants (Fig. 2c). Immunofluorescence analysis of surface PrPC further supported a correct biosynthesis and showed that all mutants are readily expressed at the plasma membrane (Fig. 2f) confirming previous results in polarized MDCK cells [61]. Of note, analysis of sPrP in media supernatants revealed that not only fl-PrP but also the truncated C1 fragment can be shed for all glycoforms (Fig. 2d; deglycosylation of supernatants for confirmation of bands is shown in Fig. 2g). Quantification (Fig. 2e) of sPrP referred to total PrP (to correct for different transfection efficiencies) showed a moderate decrease in sPrP for the monoglycosylated mutants, yet a significant reduction to ~ 50% was observed for the unglycosylated PrP-G3 (mean: 0.49 ± 0.11 compared to PrP-WT set to 1.00 ± 0.04; n = 3; ±SEM). Interestingly, as indicated by asterisks in Fig. 2a and d, for the unglycosylated PrP-G3 mutant we consistently detected a significant difference in the ratio of PrP and C1 between membrane-associated (fl-PrP: 61%; C1: 39%; ±4.5% SEM; n = 3) and shed forms (sPrP: 21%; shed C1: 79%; ±6.2%). This increase in the proportion of C1 (from 39% in lysates to 79% among the shed PrP forms) in the absence of N-glycans may relate to the longer half-life of C1 at the plasma membrane and a fast re-internalisation of unglycosylated fl-PrP [8486] which seems to be disfavored as a substrate (Fig. 2i). In contrast, for normally glycosylated PrP-WT, there appeared to be a preference for the shedding of (diglycosylated) fl-PrP over the (diglycosylated) C1 fragment when comparing fl-PrP and C1 ratios in PNGase F digested lysates (Fig. 2b) and media supernatants (Fig. 2g). This observation prompted us to investigate a potential influence of the N-terminal half of PrPC on the membrane-proximate shedding. We therefore transfected PrP-KO N2a cells with PrP-WT or with an N-terminally truncated construct (PrP-C1) corresponding to the physiological C1 fragment (Additional file 5). Despite indicating that a preference for the shedding of diglycosylated forms also exists for the C1 fragment, shedding of the latter was significantly reduced compared to fl-PrP in cells transfected with PrP-WT. Although further analysis are clearly required and differences might partially result from transient overexpression and altered surface expression of the constructs, these findings point to a role of the PrP N-terminal domain in the C-terminal shedding event. Thus, the glycosylation state as well as proteolytic truncation seem to affect PrPC shedding.

Shedding is also affected by pharmacological modulation of PrPC glycosylation

To support our findings obtained with PrP glycomutants (Fig. 2), we pharmacologically manipulated glycosylation in wild-type N2a cells. While the antibiotic tunicamycin (TM) inhibits N-glycosylation, the alkaloid swainsonine (SWA) is a known inhibitor of the further maturation of N-linked glycan structures resulting in non-mature high-mannose glycans. Treatment of cells with TM completely prevented N-glycosylation of PrPC, whereas treatment with SWA resulted in a shift in the molecular weight of diglycosylated PrPC indicating immature glycosylation (Fig. 3a). Digestion of lysates with Endo H revealed that SWA (partially) impaired complex glycosylation as shown by an altered glycopattern compared to controls (Fig. 3b). Further confirmation for the TM- and SWA-treatments and the enzymatic deglycosylation reactions is presented in a side-by-side comparison (Additional file 6). However, it should be noted that we did not reach a complete deglycosylation in the case of Endo H-digestion of lysates of SWA-treated cells. Presence of residual complex glycosylated PrPC suggests that the incubation with SWA (8 h) was too short or that Endo H digestion was incomplete. Independent of the type of treatment, PrPC was expressed at the cellular surface (Fig. 3c). Intriguingly, PrPC shedding was almost completely abolished in cells treated with TM (mean: 0.07 ± 0.03; n = 3; SEM) and significantly reduced upon treatment with SWA (0.41 ± 0.10) compared to untreated controls (set to 1.00 ± 0.12) (Fig. 3d, e). Even though only little non-mature diglycosylated PrPC was present in SWA-treated cells (Fig. 3b), this fraction seems to be the only relevant substrate for shedding. Again, these data support our previous findings (Figs. 1 and 2) showing that diglycosylated PrP is the preferential substrate for ADAM10 at the cell surface and that altered glycosylation influences PrP shedding efficiency.

Membrane anchorage and topology of PrPC determine its shedding efficiency

An additional modification that influences membrane topology, biological functions and pathophysiological roles of the prion protein is the type of membrane attachment. With PrPC being one of only few GPI-anchored substrates of ADAM10 [47, 87], we wondered how altered attachment and topology at the membrane would affect its shedding. To this end, we used mutants of PrPC either comprising a transmembrane domain instead of the GPI-anchor (PrP-TM) or carrying the GPI-anchor signal sequence (and –as a likely consequence– the GPI-anchor [61] (Puig et al. submitted)) of Thy-1, a protein described to reside in the dense cores of detergent-resistant membranes (herein referred to as lipid rafts) [88]. Whereas interaction between ADAM10 and PrPC is thought to occur at the interface between lipid rafts and non-raft regions (Fig. 4a), previous studies have shown that PrP-TM is relocated outside of rafts and turned signaling-incompetent [38, 63, 89] while PrPGPIThy-1 remains in rafts yet therein shows a different localization than PrP-WT [61] (Puig et al. submitted).
Immunofluorescent stainings of non-permeabilized cells showed surface expression (Fig. 4b) while western blot analysis revealed comparable expression of all constructs transfected into PrP-KO N2a cells (Fig. 4c). Deglycosylation of samples showed that all PrP mutants are subject to α-cleavage and confirmed an increase in molecular weight for PrP-TM due to its transmembrane domain (Fig. 4d). No alterations in the banding pattern were observed upon treatment with Endo H indicating correct glycosylation and –again– surface transport of all mutants (Fig. 4e). Lack of signal in the media for PrP-TM upon incubation of cells with PI-PLC proved the absence of a GPI-anchor and its attachment via a transmembrane domain (Fig. 4f).
Of note, despite a conserved shedding site in all constructs, shedding was completely abolished for PrP-TM (mean: 0 ± 0.02; n = 3; SEM) and significantly reduced for PrPGPIThy-1 (0.33 ± 0.06) compared to PrP-WT (set to 1.00 ± 0.05) (Fig. 4g,h). In conclusion, altered membrane attachment and, hence, changed membrane localization severely impact on PrPC shedding.

Shedding is part of a compensatory cellular network regulating PrPC homeostasis

As already shown in Fig. 1f, we consistently observed an increased cellular release of PrP via alternative routes whenever the proteolytic shedding was impaired (e.g. by inhibition of ADAM10 with GI; Fig. 5a). Since PrPC is released via microvesicles (e.g. exosomes [72, 90, 91]), we wondered whether this mechanism could compensate for abolished shedding. Inhibition of ADAM10 with GI resulted in a significant rise of exosome release (mean: 1.48 ± 0.09; compared to controls set to 1.00 ± 0.09; n = 4; SEM; Fig. 5b) without affecting the typical size of these vesicles (mean: 128 ± 12 nm; compared to controls 115 ± 2 nm; n = 4; SEM; Fig. 5c; details for characterization of exosomes are shown in Additional file 7). When amounts of exosomes were normalized, we found more than a twofold increase in their average PrPC load upon GI treatment (2.12 ± 0.08; compared to controls set to 1.00 ± 0.08; n = 3; SEM; Fig. 5d,e). PrPC levels in the corresponding cells were only moderately, yet significantly increased (1.40 ± 0.07; compared to controls set to 1.00 ± 0.07; n = 3; SEM; Fig. 5d,f). Thus, as a consequence of impaired shedding, more PrPC is packed into exosomes and more exosomes are released by N2a cells. Interestingly, such an alternative release of PrPC was not observed in murine fibroblasts (MEF) lacking ADAM10 (Additional file 8) [71] which instead accumulated PrPC to 3-fold the amount of wild-type MEFs (ADAM10 KO: 3.07 ± 0.12; WT set to 1.00 ± 0.12; n = 8; SEM). This may indicate that not all cell types possess the compensatory machinery ensuring PrPC release and membrane homeostasis.
Finally, we investigated how degradation, as a third aspect involved in cellular PrPC homeostasis, influences PrPC shedding. We blocked lysosomal degradation by treatment of N2a cells with leupeptin. As expected, cytosolic proteins (e.g. β-actin, β-tubulin, β-catenin) known to be degraded by the proteasome rather than in lysosomes were not accumulated (Fig. 5g). Instead, increased secretion of sAPPα, the proteolytic fragment of APP, indicated successful lysosomal inhibition (Fig. 5g). Of note, cell-associated PrPC levels remained rather stable despite this treatment (Leupt.: 0.97 ± 0.05; untr. Cells set to 1.00 ± 0.05; n = 3; SEM; Fig. 5g,h) yet shedding of PrPC was significantly increased (Leupt.: 2.31 ± 0.24; untr. Cells set to 1.00 ± 0.24; n = 3; SEM; Fig. 5g,i). No obvious differences in alternatively released PrP (Fig. 5g) suggests that, in this condition, shedding is the main contributor avoiding increased cellular PrPC levels.
Impaired lysosomal degradation and increased PrPC levels have recently been shown in mice lacking the sorting receptor sortilin-1 [69]. As a consequence of hindered transport to lysosomes, these mice had shown increased PrPSc conversion and shortened survival when infected with prions. Given the increase in shedding upon lysosomal inhibition with leupeptin in cells shown before, we asked whether shedding of PrPC is likewise affected by the impaired degradation due to lack of sortilin-dependent transport in vivo. In fact, we found an approximately 2-fold increase for sPrP in sortilin1-deficient mice compared to controls (Sort1 KO: 1.85 ± 0.32; Sort1 WT set to 1.00 ± 0.09; n = 4; SD; Fig. 5j). Rather than from up-regulation of sheddase activity, this increase seems to result from elevated cellular PrPC levels caused by impaired degradation (Additional file 9). Nevertheless, this demonstrates the capability of ADAM10 to release increased amounts of substrate.
In summary, these data suggest ADAM10-mediated shedding as a relevant factor regulating PrPC levels. Shedding, exosomal release and degradation of PrPC may be interconnected mechanisms that act in a compensatory manner ensuring PrPC homeostasis and allowing –if at all– only subtle changes thereof.

Discussion

Evolutionary conserved proteolytic processing of the prion protein has been described a quarter of a century ago [9294] (reviewed in [28]). However, we are just beginning to appreciate the physiological and pathological relevance of such cleavage events, which is partially due to technical difficulties in reliable detection of the respective fragments. We here present a novel antibody that detects shed PrP with high specificity and sensitivity in different applications. Despite the existence of several valuable antibodies against various epitopes in PrPC (e.g. the POM antibodies used in this study [73]), until now it has only been possible to detect shed PrP upon rather labor-intensive and error-prone immunoprecipitation from or strong concentration of cell culture supernatants [47, 48]. And even that way, contribution of PrPC released from cells by other routes (e.g. via exosomes) [91] to respective signals has to be considered. In tissue samples it has so far been impossible to specifically detect shed PrP due to the excess amounts of cell- or extracellular vesicle-associated fl-PrPC of similar molecular weight masking any signal coming from the fraction of proteolytically shed PrP. This resulted in a lack of in vivo insight. These problems have been overcome and novel findings have been made with the new antibody.
Despite confirming antibody specificity, the absence of shed PrP in forebrain homogenates from mice with a depletion of ADAM10 in forebrain neurons to our surprise indicates that, at least under physiological conditions, no other cell types in the brain contribute to shedding in a detectable manner. It also questions a shedding of (neuronal) PrPC in trans (e.g. by adjacent glia cells not depleted of the protease), a mechanism that has been shown for the ADAM10 substrate ephrin in HEK cells [95]. Our findings of abolished shedding in the absence of ADAM10 or upon pharmacological inhibition of ADAM10 also indicate that no other protease compensates for these manipulations in vitro or in vivo. Further support comes from mice coexpressing dominant negative ADAM10 with endogenous ADAM10, where we found a comparably strong (~ 50%) reduction in PrP shedding. Instead, previous western blot analyses of sAPPα in ADAM10 d.n. mice only showed a reduction of ~ 25% [96, 97] hinting at the known contribution of ADAM17/TACE in the cleavage of APP [98]. It should be considered that cleavage by another protease at a slightly different cleavage site would prevent detection with our antibody. However, our previous results obtained by pull-down of shed PrP from media of primary ADAM10 knockout neurons with classical PrP antibodies [48], together with a recent biophysical study [31], and the lack of any other reported candidate protease linked to the membrane-proximate shedding of PrPC, support the view of ADAM10 as the only relevant sheddase of PrPC. This is in clear contrast to the cleavage of other typical ADAM10 substrates such as APP, which –as mentioned above– to varying degrees and dependent on the experimental paradigm, can also be processed by other proteases [71, 99101].
Our analysis suggests that diglycosylated PrPC is the preferred glycoform to be shed by ADAM10, whereas mono- and especially unglycosylated forms seem to be relatively disfavored. Our data also indicates that this finding not simply results from differences in the availability of individual glycoforms as substrates at the plasma membrane under normal conditions (our transfected glycomutants as well as PrPC in cells treated with TM or SWA were by all means localized at the surface). An alternative explanation could be that shed diglycosylated PrP is more protected than the other shed forms from potential cellular uptake and degradation and, thus, more abundant. In any case, among all soluble PrPC fragments released from the cell by the proteolytic cleavages described to date, shed PrP is the only one that is glycosylated. As discussed earlier [28] this may well impact its binding affinities to both, toxic extracellular oligomers as well as physiological binding partners (e.g. surface signaling receptors), and thus define its specific biological functions. Moreover, by the predominantly diglycosylated state, physiologically shed PrP clearly differs from anchorless, mainly unglycosylated PrP of transgenic mice used in several seminal prion inoculation studies in the past [54, 102104]. This difference has to be considered and, in the context of prion diseases, might explain why transgenic anchorless PrP efficiently converts to PrPSc and can even spontaneously form prions [54, 103], whereas shed PrP rather seems to block PrPSc formation in mice [50]. Fittingly, the N-glycans are known to influence transmissibility and conversion to PrPSc [62, 8083].
Altered shedding efficiency for different glycoforms, however, might in part also be caused by a different sorting given that the glycans have a significant impact on the polarized trafficking of PrPC in MDCK cells [61]. Despite a role for the N-glycans, we also demonstrated that changes in the type of membrane anchorage and, as a likely consequence, altered membrane topology affects shedding. Shifting PrPC outside of rafts by addition of a transmembrane domain [63, 89] completely abolished the shedding while the assumed re-localization of PrPC within lipid rafts via exchange of the GPI-anchor signal sequence [61, 88] reduced shedding to ~ 30% in N2a cells. The latter is in good agreement with unpublished data obtained in transgenic mice expressing the same PrPGPIThy-1 construct (Puig et al., submitted). Instead of changing the anchorage of the substrate as done here, others have changed membrane attachment of the protease [105]. Lipid raft targeting of ADAM10 by addition of a GPI-anchor in that study severely affected APP processing. Unfortunately, processing of PrPC was not investigated there.
Since ADAM10 is mainly located outside of lipid rafts [105, 106], whereas PrPC is a resident of these microdomains, transient interaction between protease and substrate (presumably regulated by accessory proteins such as tetraspanins [107109]) and cleavage is thought to occur at the periphery of rafts. This molecular get-together might further be supported by the capacity of PrPC to leave and re-enter lipid rafts in a constitutive manner [85, 110, 111]. Our findings also suggest an impact of the flexible N-terminal part of PrPC on the shedding efficiency. Whether this unanticipated influence is due to sterical aspects or rather reflects the role of regulatory binding partners known to especially interact with the N-terminal half of PrPC [5, 27, 112], deserves further investigations.
Our data indicate that shedding is a relevant mechanism embedded in a compensatory machinery ensuring homeostasis of PrPC. In neurons and neuronal cells, this system (involving proteolytic and exosomal release as well as trafficking to lysosomes) seems to ensure that cell-associated PrPC levels are kept stable or –at most– increase twofold upon perturbation (as indicated in some experiments of this study and observed in neurons or mice lacking the sheddase ADAM10 [48, 50] or the transport factor sortilin-1 [69]). Interestingly, a recent study showed that exosomal release is controlled by PrPC membrane levels [113]. Though clearly requiring further investigation, it might be speculated that other cell types, such as fibroblasts studied here, do not possess the system to compensate for such perturbation in one of the mechanisms discussed above, and consequently accumulate PrPC to higher levels.
Manipulation of PrPC shedding is feasible and might be of therapeutic interest. Despite the challenge by possible side effects due to the broad spectrum of ADAM10 substrates, one obvious question then is into which direction to modify PrPC shedding [87, 114].
With regard to neurodegenerative proteinopathies, such as Alzheimer’s or prion diseases, stimulation of this cleavage will likely be beneficial. First, it reduces PrPC levels at the cell surface and may thereby lower neurotoxicity. Moreover, several studies showed that soluble PrP targets toxic oligomers and fibrils in the extracellular space [5558, 115]. In prion diseases, shedding efficiency inversely correlates with PrPSc formation [50, 52]. Notably, resveratrol, the drug that was used here to stimulate shedding, reduced PrPSc formation and prion infectivity in a recent study [116]. Whether this anti-prion efficacy is indeed related to shedding, remains to be investigated. Besides proteinopathies, positive effects of stimulated shedding can also be expected given the potential role of this fragment in neurite outgrowth [13, 14] and neuroprotection [15, 49]. In that way, the role of shed PrP is reminiscent of sAPPα, the APP-derived fragment also generated by ADAM10 [117].
Other pathological conditions, in contrast, may rather require inhibition of PrPC shedding: it is intriguing that both, ADAM10 [118, 119] and PrPC, have been linked with immune signaling and chronic inflammatory processes [120, 121] as well as with tumorigenesis and cancer progression [122124], where expression levels of these two proteins generally correlate with poor prognosis. Though this could well be unrelated co-incidence, it might also be speculated that these pathophysiological roles are partially related to the production of shed PrP. Of note, it is precisely shed PrP that was causally linked with chronic inflammatory neuropathology in HIV patients [60] and development of tumours in the central nervous system [59] in two recent studies. This further supports the relevance of shed PrP in different pathophysiological conditions and highlights the need for further studies on the ADAM10-mediated shedding of PrPC.

Conclusion

Proteolytic shedding of the prion protein has most recently attracted scientific interest with regard to diverse pathological conditions affecting the brain. Using a novel antibody for the specific detection of shed PrP, we demonstrated structural and regulatory aspects influencing this cleavage and show that it can - in principle - be pharmacologically manipulated. The latter, together with the rather ubiquitous expression of PrPC in several tissues and cell lines, as well as the lack of compensation by other proteases discussed above, also turns (i) PrPC into an ideal “control” substrate, (ii) assessment of PrPC shedding into a reliable “read out”, and (iii) our antibody into a valuable tool for any future studies investigating ADAM10-mediated cleavages and their pharmacological accessibility. With direct regard to the shedding of PrPC, both, therapeutic stimulation as well as inhibition, may be conceivable depending on the pathological context.

Acknowledgements

We thank Dr. Dirk Kamin (Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany), Kristin Hartmann (Mouse Pathology Core Unit, UKE, Hamburg, Germany) and the UKE Microscopy Imaging Facility (umif) for technical support. We apologize to all colleagues whose important contributions to this field could not be cited due to space limitations.

Funding

We are thankful for support by the Deutsche Forschungsgemeinschaft (DFG) Sonderforschungsbereich 877 (projects A12 (to HCA, PS and MG) and A3 (to PS)), the Creutzfeldt-Jakob Disease Foundation, Inc. (to HCA) and the Werner-Otto-Stiftung (to LL, BP and HCA).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request. The authors will be happy to provide the sPrPG228 antibody characterized herein as research tool on reasonable request.

Authors’ information

Not applicable.

Ethics approval

No experiments on living animals were conducted for this study. However, housing and sacrification of animals as well as use of animal material in this study was in strict compliance with the Guide for the Care and Use of Laboratory Animals and ethics guidelines of the responsible local authorities.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Additional file 2: (.jpg) Species specificity of the sPrPG228 antibody. (A) Comparison of the C-terminal amino acid sequence of PrPC in mouse (Mus musculus), rat (Rattus norvegicus) and rabbit (Oryctolagus cuniculus) (source: www.​uniprot.​org). “NH 2 -…-” indicates the N-terminal direction, “-GPI” the C-terminal GPI-anchor attachment site. Asterisks indicate position of ADAM10-mediated shedding in mice and rats with Gly228 representing the new C-terminus of shed PrP. Note the sequence difference compared with rabbit PrPC. (B) Western blot analysis of forebrain homogenates from different murine models (tga20, Prnp0/0, wild-type (C57BL/6)) as well as from rat and rabbit. As expected for its epitope, the sPrPG228 antibody detects sPrP in mouse (tga20 and wild-type) and rat, whereas the brain sample of rabbit only presents an immunoglobulin light chain (rb Ig-LC) signal at 25 kDa resulting from the anti-rabbit secondary antibody used for detection. Re-probing the blot with POM2 antibody reveals expression levels of PrPC. (JPEG 388 kb)
Additional file 6: (.jpg) Side-by-side comparison of SWA and TM treatments and enzymatic deglycosylation reactions. Western blot of untreated (untr.), SWA- or TM-treated N2a cells showing lysates without (−) or with (+) enzymatic treatment for differential deglycosylation (Endo H or PNGase F). As also shown in Fig. 3a and b, TM-treatment causes a complete inhibition of PrP glycosylation, whereas SWA-treatment results in a shift in the banding pattern (compared to untreated cells) and (at least partial) Endo H sensitivity due to inhibition of complex glycosylation. Changes in the glycopattern and running behaviour support the functioning of our enzymatic deglycosylation protocols also performed for the experiments shown in Figs. 2, 3 and 4. Actin is shown as a loading control. (JPEG 221 kb)
Additional file 9: (.jpg) Quantification of fl-PrP levels and ratio of sPrP/fl-PrP in Sort1 knockout mice. These quantifications refer to main Fig. 5j. (A) Increased amounts of fl-PrP are found in brains of Sort1 KO mice (2.11 ± 0.23; p = 0.0004; n = 4) compared to controls (WT set to 1.00 ± 0.21; SD). Actin served as loading control and for reference in densitometric quantification. (B) No significant differences are detected in the ratio of sPrP to fl-PrP between Sort1 KO mouse brains (0.85 ± 0.07, p = 0.128; n = 4) and controls (WT set to 1.00 ± 0.14). (JPEG 229 kb)
Literatur
1.
Zurück zum Zitat Seidah NG, et al. The multifaceted proprotein convertases: their unique, redundant, complementary, and opposite functions. J Biol Chem. 2013;288(30):21473–81.PubMedPubMedCentralCrossRef Seidah NG, et al. The multifaceted proprotein convertases: their unique, redundant, complementary, and opposite functions. J Biol Chem. 2013;288(30):21473–81.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Aguzzi A, Baumann F, Bremer J. The prion's elusive reason for being. Annu Rev Neurosci. 2008;31:439–77.PubMedCrossRef Aguzzi A, Baumann F, Bremer J. The prion's elusive reason for being. Annu Rev Neurosci. 2008;31:439–77.PubMedCrossRef
4.
6.
Zurück zum Zitat Steele AD, et al. Prion protein (PrPc) positively regulates neural precursor proliferation during developmental and adult mammalian neurogenesis. Proc Natl Acad Sci U S A. 2006;103(9):3416–21.PubMedPubMedCentralCrossRef Steele AD, et al. Prion protein (PrPc) positively regulates neural precursor proliferation during developmental and adult mammalian neurogenesis. Proc Natl Acad Sci U S A. 2006;103(9):3416–21.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Malaga-Trillo E, et al. Regulation of embryonic cell adhesion by the prion protein. PLoS Biol. 2009;7(3):e55.PubMedCrossRef Malaga-Trillo E, et al. Regulation of embryonic cell adhesion by the prion protein. PLoS Biol. 2009;7(3):e55.PubMedCrossRef
9.
Zurück zum Zitat Schmitt-Ulms G, et al. Binding of neural cell adhesion molecules (N-CAMs) to the cellular prion protein. J Mol Biol. 2001;314(5):1209–25.PubMedCrossRef Schmitt-Ulms G, et al. Binding of neural cell adhesion molecules (N-CAMs) to the cellular prion protein. J Mol Biol. 2001;314(5):1209–25.PubMedCrossRef
10.
Zurück zum Zitat Graner E, et al. Cellular prion protein binds laminin and mediates neuritogenesis. Brain Res Mol Brain Res. 2000;76(1):85–92.PubMedCrossRef Graner E, et al. Cellular prion protein binds laminin and mediates neuritogenesis. Brain Res Mol Brain Res. 2000;76(1):85–92.PubMedCrossRef
11.
Zurück zum Zitat Chen S, et al. Prion protein as trans-interacting partner for neurons is involved in neurite outgrowth and neuronal survival. Mol Cell Neurosci. 2003;22(2):227–33.PubMedCrossRef Chen S, et al. Prion protein as trans-interacting partner for neurons is involved in neurite outgrowth and neuronal survival. Mol Cell Neurosci. 2003;22(2):227–33.PubMedCrossRef
12.
Zurück zum Zitat Santuccione A, et al. Prion protein recruits its neuronal receptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth. J Cell Biol. 2005;169(2):341–54.PubMedPubMedCentralCrossRef Santuccione A, et al. Prion protein recruits its neuronal receptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth. J Cell Biol. 2005;169(2):341–54.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Kanaani J, et al. Recombinant prion protein induces rapid polarization and development of synapses in embryonic rat hippocampal neurons in vitro. J Neurochem. 2005;95(5):1373–86.PubMedCrossRef Kanaani J, et al. Recombinant prion protein induces rapid polarization and development of synapses in embryonic rat hippocampal neurons in vitro. J Neurochem. 2005;95(5):1373–86.PubMedCrossRef
14.
Zurück zum Zitat Amin L, et al. Characterization of prion protein function by focal neurite stimulation. J Cell Sci. 2016;129(20):3878–91.PubMedCrossRef Amin L, et al. Characterization of prion protein function by focal neurite stimulation. J Cell Sci. 2016;129(20):3878–91.PubMedCrossRef
15.
Zurück zum Zitat Bounhar Y, et al. Prion protein protects human neurons against Bax-mediated apoptosis. J Biol Chem. 2001;276(42):39145–9.PubMedCrossRef Bounhar Y, et al. Prion protein protects human neurons against Bax-mediated apoptosis. J Biol Chem. 2001;276(42):39145–9.PubMedCrossRef
16.
Zurück zum Zitat Roucou X, Gains M, LeBlanc AC. Neuroprotective functions of prion protein. J Neurosci Res. 2004;75(2):153–61.PubMedCrossRef Roucou X, Gains M, LeBlanc AC. Neuroprotective functions of prion protein. J Neurosci Res. 2004;75(2):153–61.PubMedCrossRef
17.
Zurück zum Zitat Guillot-Sestier MV, et al. The alpha-secretase-derived N-terminal product of cellular prion, N1, displays neuroprotective function in vitro and in vivo. J Biol Chem. 2009;284(51):35973–86.PubMedPubMedCentralCrossRef Guillot-Sestier MV, et al. The alpha-secretase-derived N-terminal product of cellular prion, N1, displays neuroprotective function in vitro and in vivo. J Biol Chem. 2009;284(51):35973–86.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Tobler I, et al. Altered circadian activity rhythms and sleep in mice devoid of prion protein. Nature. 1996;380(6575):639–42.PubMedCrossRef Tobler I, et al. Altered circadian activity rhythms and sleep in mice devoid of prion protein. Nature. 1996;380(6575):639–42.PubMedCrossRef
20.
Zurück zum Zitat Bremer J, et al. Axonal prion protein is required for peripheral myelin maintenance. Nat Neurosci. 2010;13(3):310–8.PubMedCrossRef Bremer J, et al. Axonal prion protein is required for peripheral myelin maintenance. Nat Neurosci. 2010;13(3):310–8.PubMedCrossRef
22.
Zurück zum Zitat Brown DR, et al. The cellular prion protein binds copper in vivo. Nature. 1997;390(6661):684–7.PubMedCrossRef Brown DR, et al. The cellular prion protein binds copper in vivo. Nature. 1997;390(6661):684–7.PubMedCrossRef
24.
Zurück zum Zitat Mouillet-Richard S, et al. Signal transduction through prion protein. Science. 2000;289(5486):1925–8.PubMedCrossRef Mouillet-Richard S, et al. Signal transduction through prion protein. Science. 2000;289(5486):1925–8.PubMedCrossRef
26.
Zurück zum Zitat Lewis V, Hooper NM. The role of lipid rafts in prion protein biology. Front Biosci (Landmark Ed). 2011;16:151–68.CrossRef Lewis V, Hooper NM. The role of lipid rafts in prion protein biology. Front Biosci (Landmark Ed). 2011;16:151–68.CrossRef
27.
28.
Zurück zum Zitat Linsenmeier L, et al. Diverse functions of the prion protein - does proteolytic processing hold the key? Biochim Biophys Acta. 2017; Linsenmeier L, et al. Diverse functions of the prion protein - does proteolytic processing hold the key? Biochim Biophys Acta. 2017;
29.
Zurück zum Zitat Altmeppen HC, et al. Proteolytic processing of the prion protein in health and disease. Am J Neurodegener Dis. 2012;1(1):15–31.PubMedPubMedCentral Altmeppen HC, et al. Proteolytic processing of the prion protein in health and disease. Am J Neurodegener Dis. 2012;1(1):15–31.PubMedPubMedCentral
30.
Zurück zum Zitat Mays CE, et al. Endoproteolytic processing of the mammalian prion glycoprotein family. FEBS J. 2014;281(3):862–76.PubMedCrossRef Mays CE, et al. Endoproteolytic processing of the mammalian prion glycoprotein family. FEBS J. 2014;281(3):862–76.PubMedCrossRef
31.
Zurück zum Zitat McDonald AJ, et al. A new paradigm for enzymatic control of alpha-cleavage and beta-cleavage of the prion protein. J Biol Chem. 2014;289(2):803–13.PubMedCrossRef McDonald AJ, et al. A new paradigm for enzymatic control of alpha-cleavage and beta-cleavage of the prion protein. J Biol Chem. 2014;289(2):803–13.PubMedCrossRef
32.
Zurück zum Zitat Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science. 1982;216(4542):136–44.PubMedCrossRef Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science. 1982;216(4542):136–44.PubMedCrossRef
33.
Zurück zum Zitat Aguzzi A, Falsig J. Prion propagation, toxicity and degradation. Nat Neurosci. 2012;15(7):936–9.PubMedCrossRef Aguzzi A, Falsig J. Prion propagation, toxicity and degradation. Nat Neurosci. 2012;15(7):936–9.PubMedCrossRef
34.
Zurück zum Zitat Collinge J. Mammalian prions and their wider relevance in neurodegenerative diseases. Nature. 2016;539(7628):217–26.PubMedCrossRef Collinge J. Mammalian prions and their wider relevance in neurodegenerative diseases. Nature. 2016;539(7628):217–26.PubMedCrossRef
35.
Zurück zum Zitat Basler K, et al. Scrapie and cellular PrP isoforms are encoded by the same chromosomal gene. Cell. 1986;46(3):417–28.PubMedCrossRef Basler K, et al. Scrapie and cellular PrP isoforms are encoded by the same chromosomal gene. Cell. 1986;46(3):417–28.PubMedCrossRef
38.
Zurück zum Zitat Resenberger UK, et al. The cellular prion protein mediates neurotoxic signalling of beta-sheet-rich conformers independent of prion replication. EMBO J. 2011;30(10):2057–70.PubMedPubMedCentralCrossRef Resenberger UK, et al. The cellular prion protein mediates neurotoxic signalling of beta-sheet-rich conformers independent of prion replication. EMBO J. 2011;30(10):2057–70.PubMedPubMedCentralCrossRef
39.
40.
Zurück zum Zitat Um JW, et al. Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer abeta oligomer bound to cellular prion protein. Neuron. 2013;79(5):887–902.PubMedPubMedCentralCrossRef Um JW, et al. Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer abeta oligomer bound to cellular prion protein. Neuron. 2013;79(5):887–902.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Dohler F, et al. High molecular mass assemblies of amyloid-beta oligomers bind prion protein in patients with Alzheimer's disease. Brain. 2014;137(Pt 3):873–86.PubMedCrossRef Dohler F, et al. High molecular mass assemblies of amyloid-beta oligomers bind prion protein in patients with Alzheimer's disease. Brain. 2014;137(Pt 3):873–86.PubMedCrossRef
42.
Zurück zum Zitat Haas LT, et al. Metabotropic glutamate receptor 5 couples cellular prion protein to intracellular signalling in Alzheimer's disease. Brain. 2016;139(Pt 2):526–46.PubMedCrossRef Haas LT, et al. Metabotropic glutamate receptor 5 couples cellular prion protein to intracellular signalling in Alzheimer's disease. Brain. 2016;139(Pt 2):526–46.PubMedCrossRef
43.
Zurück zum Zitat Ferreira DG, et al. alpha-synuclein interacts with PrPC to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci. 2017;20(11):1569–79.PubMedCrossRef Ferreira DG, et al. alpha-synuclein interacts with PrPC to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci. 2017;20(11):1569–79.PubMedCrossRef
44.
Zurück zum Zitat Urrea L, et al. The cellular prion protein (PrPC) as neuronal receptor for alpha-synuclein. Prion. 2017;11(4):226–33.PubMedCrossRef Urrea L, et al. The cellular prion protein (PrPC) as neuronal receptor for alpha-synuclein. Prion. 2017;11(4):226–33.PubMedCrossRef
45.
Zurück zum Zitat Altmeppen HC, et al. Roles of endoproteolytic alpha-cleavage and shedding of the prion protein in neurodegeneration. FEBS J. 2013;280(18):4338–47.PubMedCrossRef Altmeppen HC, et al. Roles of endoproteolytic alpha-cleavage and shedding of the prion protein in neurodegeneration. FEBS J. 2013;280(18):4338–47.PubMedCrossRef
46.
Zurück zum Zitat Beland M, Roucou X. Taking advantage of physiological proteolytic processing of the prion protein for a therapeutic perspective in prion and Alzheimer diseases. Prion. 2014;8(1):106–10.PubMedCrossRef Beland M, Roucou X. Taking advantage of physiological proteolytic processing of the prion protein for a therapeutic perspective in prion and Alzheimer diseases. Prion. 2014;8(1):106–10.PubMedCrossRef
47.
Zurück zum Zitat Taylor DR, et al. Role of ADAMs in the ectodomain shedding and conformational conversion of the prion protein. J Biol Chem. 2009;284(34):22590–600.PubMedPubMedCentralCrossRef Taylor DR, et al. Role of ADAMs in the ectodomain shedding and conformational conversion of the prion protein. J Biol Chem. 2009;284(34):22590–600.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Altmeppen HC, et al. Lack of a-disintegrin-and-metalloproteinase ADAM10 leads to intracellular accumulation and loss of shedding of the cellular prion protein in vivo. Mol Neurodegener. 2011;6:36.PubMedPubMedCentralCrossRef Altmeppen HC, et al. Lack of a-disintegrin-and-metalloproteinase ADAM10 leads to intracellular accumulation and loss of shedding of the cellular prion protein in vivo. Mol Neurodegener. 2011;6:36.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Race B, et al. Prion protein on astrocytes or in extracellular fluid impedes neurodegeneration induced by truncated prion protein. Exp Neurol. 2009;217(2):347–52.PubMedPubMedCentralCrossRef Race B, et al. Prion protein on astrocytes or in extracellular fluid impedes neurodegeneration induced by truncated prion protein. Exp Neurol. 2009;217(2):347–52.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Altmeppen HC, et al. The sheddase ADAM10 is a potent modulator of prion disease. elife. 2015;4 Altmeppen HC, et al. The sheddase ADAM10 is a potent modulator of prion disease. elife. 2015;4
52.
Zurück zum Zitat Endres K, et al. Influence of ADAM10 on prion protein processing and scrapie infectiosity in vivo. Neurobiol Dis. 2009;36(2):233–41.PubMedCrossRef Endres K, et al. Influence of ADAM10 on prion protein processing and scrapie infectiosity in vivo. Neurobiol Dis. 2009;36(2):233–41.PubMedCrossRef
53.
Zurück zum Zitat Meier P, et al. Soluble dimeric prion protein binds PrP(Sc) in vivo and antagonizes prion disease. Cell. 2003;113(1):49–60.PubMedCrossRef Meier P, et al. Soluble dimeric prion protein binds PrP(Sc) in vivo and antagonizes prion disease. Cell. 2003;113(1):49–60.PubMedCrossRef
54.
Zurück zum Zitat Chesebro B, et al. Anchorless prion protein results in infectious amyloid disease without clinical scrapie. Science. 2005;308(5727):1435–9.PubMedCrossRef Chesebro B, et al. Anchorless prion protein results in infectious amyloid disease without clinical scrapie. Science. 2005;308(5727):1435–9.PubMedCrossRef
56.
57.
Zurück zum Zitat Fluharty BR, et al. An N-terminal fragment of the prion protein binds to amyloid-beta oligomers and inhibits their neurotoxicity in vivo. J Biol Chem. 2013;288(11):7857–66.PubMedPubMedCentralCrossRef Fluharty BR, et al. An N-terminal fragment of the prion protein binds to amyloid-beta oligomers and inhibits their neurotoxicity in vivo. J Biol Chem. 2013;288(11):7857–66.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Scott-McKean JJ, et al. Soluble prion protein and its N-terminal fragment prevent impairment of synaptic plasticity by Abeta oligomers: implications for novel therapeutic strategy in Alzheimer's disease. Neurobiol Dis. 2016;91:124–31.PubMedPubMedCentralCrossRef Scott-McKean JJ, et al. Soluble prion protein and its N-terminal fragment prevent impairment of synaptic plasticity by Abeta oligomers: implications for novel therapeutic strategy in Alzheimer's disease. Neurobiol Dis. 2016;91:124–31.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Provenzano L, et al. Cellular prion protein (PrPC) in the development of Merlin-deficient tumours. Oncogene. 2017; Provenzano L, et al. Cellular prion protein (PrPC) in the development of Merlin-deficient tumours. Oncogene. 2017;
60.
Zurück zum Zitat Megra BW, Eugenin EA, Berman JW. The role of shed PrPc in the Neuropathogenesis of HIV infection. J Immunol. 2017;199(1):224–32.PubMedCrossRef Megra BW, Eugenin EA, Berman JW. The role of shed PrPc in the Neuropathogenesis of HIV infection. J Immunol. 2017;199(1):224–32.PubMedCrossRef
61.
Zurück zum Zitat Puig B, et al. N-glycans and glycosylphosphatidylinositol-anchor act on polarized sorting of mouse PrP(C) in Madin-Darby canine kidney cells. PLoS One. 2011;6(9):e24624.PubMedPubMedCentralCrossRef Puig B, et al. N-glycans and glycosylphosphatidylinositol-anchor act on polarized sorting of mouse PrP(C) in Madin-Darby canine kidney cells. PLoS One. 2011;6(9):e24624.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Winklhofer KF, et al. Inhibition of complex glycosylation increases the formation of PrPsc. Traffic. 2003;4(5):313–22.PubMedCrossRef Winklhofer KF, et al. Inhibition of complex glycosylation increases the formation of PrPsc. Traffic. 2003;4(5):313–22.PubMedCrossRef
63.
Zurück zum Zitat Taraboulos A, et al. Cholesterol depletion and modification of COOH-terminal targeting sequence of the prion protein inhibit formation of the scrapie isoform. J Cell Biol. 1995;129(1):121–32.PubMedCrossRef Taraboulos A, et al. Cholesterol depletion and modification of COOH-terminal targeting sequence of the prion protein inhibit formation of the scrapie isoform. J Cell Biol. 1995;129(1):121–32.PubMedCrossRef
64.
Zurück zum Zitat Kascsak RJ, et al. Mouse polyclonal and monoclonal antibody to scrapie-associated fibril proteins. J Virol. 1987;61(12):3688–93.PubMedPubMedCentral Kascsak RJ, et al. Mouse polyclonal and monoclonal antibody to scrapie-associated fibril proteins. J Virol. 1987;61(12):3688–93.PubMedPubMedCentral
65.
Zurück zum Zitat Bueler H, et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature. 1992;356(6370):577–82.PubMedCrossRef Bueler H, et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature. 1992;356(6370):577–82.PubMedCrossRef
66.
Zurück zum Zitat Fischer M, et al. Prion protein (PrP) with amino-proximal deletions restoring susceptibility of PrP knockout mice to scrapie. EMBO J. 1996;15(6):1255–64.PubMedPubMedCentral Fischer M, et al. Prion protein (PrP) with amino-proximal deletions restoring susceptibility of PrP knockout mice to scrapie. EMBO J. 1996;15(6):1255–64.PubMedPubMedCentral
67.
Zurück zum Zitat Prox, J., et al., Postnatal disruption of the disintegrin/metalloproteinase ADAM10 in brain causes epileptic seizures, learning deficits, altered spine morphology, and defective synaptic functions. J Neurosci, 2013. 33(32): p. 12915–12928, 12928a. Prox, J., et al., Postnatal disruption of the disintegrin/metalloproteinase ADAM10 in brain causes epileptic seizures, learning deficits, altered spine morphology, and defective synaptic functions. J Neurosci, 2013. 33(32): p. 12915–12928, 12928a.
68.
Zurück zum Zitat Postina R, et al. A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J Clin Invest. 2004;113(10):1456–64.PubMedPubMedCentralCrossRef Postina R, et al. A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J Clin Invest. 2004;113(10):1456–64.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Hartmann D, et al. The disintegrin/metalloprotease ADAM 10 is essential for notch signalling but not for alpha-secretase activity in fibroblasts. Hum Mol Genet. 2002;11(21):2615–24.PubMedCrossRef Hartmann D, et al. The disintegrin/metalloprotease ADAM 10 is essential for notch signalling but not for alpha-secretase activity in fibroblasts. Hum Mol Genet. 2002;11(21):2615–24.PubMedCrossRef
72.
Zurück zum Zitat Falker C, et al. Exosomal cellular prion protein drives fibrillization of amyloid beta and counteracts amyloid beta-mediated neurotoxicity. J Neurochem. 2016;137(1):88–100.PubMedCrossRef Falker C, et al. Exosomal cellular prion protein drives fibrillization of amyloid beta and counteracts amyloid beta-mediated neurotoxicity. J Neurochem. 2016;137(1):88–100.PubMedCrossRef
73.
Zurück zum Zitat Polymenidou M, et al. The POM monoclonals: a comprehensive set of antibodies to non-overlapping prion protein epitopes. PLoS One. 2008;3(12):e3872.PubMedPubMedCentralCrossRef Polymenidou M, et al. The POM monoclonals: a comprehensive set of antibodies to non-overlapping prion protein epitopes. PLoS One. 2008;3(12):e3872.PubMedPubMedCentralCrossRef
74.
75.
Zurück zum Zitat Holthoewer D, et al. Acitretin, an enhancer of alpha-secretase expression, crosses the blood-brain barrier and is not eliminated by P-glycoprotein. Neurodegener Dis. 2012;10(1–4):224–8.PubMedCrossRef Holthoewer D, et al. Acitretin, an enhancer of alpha-secretase expression, crosses the blood-brain barrier and is not eliminated by P-glycoprotein. Neurodegener Dis. 2012;10(1–4):224–8.PubMedCrossRef
76.
Zurück zum Zitat Sathya M, et al. Resveratrol intervenes cholesterol- and isoprenoid-mediated Amyloidogenic processing of AbetaPP in familial Alzheimer's disease. J Alzheimers Dis. 2017;60(s1):S3–S23.PubMedCrossRef Sathya M, et al. Resveratrol intervenes cholesterol- and isoprenoid-mediated Amyloidogenic processing of AbetaPP in familial Alzheimer's disease. J Alzheimers Dis. 2017;60(s1):S3–S23.PubMedCrossRef
77.
Zurück zum Zitat Hundhausen C, et al. The disintegrin-like metalloproteinase ADAM10 is involved in constitutive cleavage of CX3CL1 (fractalkine) and regulates CX3CL1-mediated cell-cell adhesion. Blood. 2003;102(4):1186–95.PubMedCrossRef Hundhausen C, et al. The disintegrin-like metalloproteinase ADAM10 is involved in constitutive cleavage of CX3CL1 (fractalkine) and regulates CX3CL1-mediated cell-cell adhesion. Blood. 2003;102(4):1186–95.PubMedCrossRef
79.
Zurück zum Zitat Walmsley AR, et al. Alpha-cleavage of the prion protein occurs in a late compartment of the secretory pathway and is independent of lipid rafts. Mol Cell Neurosci. 2009;40(2):242–8.PubMedCrossRef Walmsley AR, et al. Alpha-cleavage of the prion protein occurs in a late compartment of the secretory pathway and is independent of lipid rafts. Mol Cell Neurosci. 2009;40(2):242–8.PubMedCrossRef
80.
Zurück zum Zitat Korth C, Kaneko K, Prusiner SB. Expression of unglycosylated mutated prion protein facilitates PrP(Sc) formation in neuroblastoma cells infected with different prion strains. J Gen Virol. 2000;81(Pt 10):2555–63.PubMedCrossRef Korth C, Kaneko K, Prusiner SB. Expression of unglycosylated mutated prion protein facilitates PrP(Sc) formation in neuroblastoma cells infected with different prion strains. J Gen Virol. 2000;81(Pt 10):2555–63.PubMedCrossRef
81.
Zurück zum Zitat Neuendorf E, et al. Glycosylation deficiency at either one of the two glycan attachment sites of cellular prion protein preserves susceptibility to bovine spongiform encephalopathy and scrapie infections. J Biol Chem. 2004;279(51):53306–16.PubMedCrossRef Neuendorf E, et al. Glycosylation deficiency at either one of the two glycan attachment sites of cellular prion protein preserves susceptibility to bovine spongiform encephalopathy and scrapie infections. J Biol Chem. 2004;279(51):53306–16.PubMedCrossRef
82.
Zurück zum Zitat Katorcha E, et al. Sialylation of prion protein controls the rate of prion amplification, the cross-species barrier, the ratio of PrPSc glycoform and prion infectivity. PLoS Pathog. 2014;10(9):e1004366.PubMedPubMedCentralCrossRef Katorcha E, et al. Sialylation of prion protein controls the rate of prion amplification, the cross-species barrier, the ratio of PrPSc glycoform and prion infectivity. PLoS Pathog. 2014;10(9):e1004366.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Wiseman FK, et al. The glycosylation status of PrPC is a key factor in determining transmissible spongiform encephalopathy transmission between species. J Virol. 2015;89(9):4738–47.PubMedPubMedCentralCrossRef Wiseman FK, et al. The glycosylation status of PrPC is a key factor in determining transmissible spongiform encephalopathy transmission between species. J Virol. 2015;89(9):4738–47.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Shyng SL, Huber MT, Harris DA. A prion protein cycles between the cell surface and an endocytic compartment in cultured neuroblastoma cells. J Biol Chem. 1993;268(21):15922–8.PubMed Shyng SL, Huber MT, Harris DA. A prion protein cycles between the cell surface and an endocytic compartment in cultured neuroblastoma cells. J Biol Chem. 1993;268(21):15922–8.PubMed
85.
86.
Zurück zum Zitat Nunziante M, Gilch S, Schatzl HM. Essential role of the prion protein N terminus in subcellular trafficking and half-life of cellular prion protein. J Biol Chem. 2003;278(6):3726–34.PubMedCrossRef Nunziante M, Gilch S, Schatzl HM. Essential role of the prion protein N terminus in subcellular trafficking and half-life of cellular prion protein. J Biol Chem. 2003;278(6):3726–34.PubMedCrossRef
87.
Zurück zum Zitat Saftig P, Lichtenthaler SF. The alpha secretase ADAM10: a metalloprotease with multiple functions in the brain. Prog Neurobiol. 2015; Saftig P, Lichtenthaler SF. The alpha secretase ADAM10: a metalloprotease with multiple functions in the brain. Prog Neurobiol. 2015;
88.
92.
Zurück zum Zitat Harris DA, et al. Processing of a cellular prion protein: identification of N- and C-terminal cleavage sites. Biochemistry. 1993;32(4):1009–16.PubMedCrossRef Harris DA, et al. Processing of a cellular prion protein: identification of N- and C-terminal cleavage sites. Biochemistry. 1993;32(4):1009–16.PubMedCrossRef
93.
Zurück zum Zitat Borchelt DR, et al. Release of the cellular prion protein from cultured cells after loss of its glycoinositol phospholipid anchor. Glycobiology. 1993;3(4):319–29.PubMedCrossRef Borchelt DR, et al. Release of the cellular prion protein from cultured cells after loss of its glycoinositol phospholipid anchor. Glycobiology. 1993;3(4):319–29.PubMedCrossRef
94.
Zurück zum Zitat Chen SG, et al. Truncated forms of the human prion protein in normal brain and in prion diseases. J Biol Chem. 1995;270(32):19173–80.PubMedCrossRef Chen SG, et al. Truncated forms of the human prion protein in normal brain and in prion diseases. J Biol Chem. 1995;270(32):19173–80.PubMedCrossRef
95.
Zurück zum Zitat Janes PW, et al. Adam meets Eph: an ADAM substrate recognition module acts as a molecular switch for ephrin cleavage in trans. Cell. 2005;123(2):291–304.PubMedCrossRef Janes PW, et al. Adam meets Eph: an ADAM substrate recognition module acts as a molecular switch for ephrin cleavage in trans. Cell. 2005;123(2):291–304.PubMedCrossRef
96.
Zurück zum Zitat Clement AB, et al. Effects of neuron-specific ADAM10 modulation in an in vivo model of acute excitotoxic stress. Neuroscience. 2008;152(2):459–68.PubMedCrossRef Clement AB, et al. Effects of neuron-specific ADAM10 modulation in an in vivo model of acute excitotoxic stress. Neuroscience. 2008;152(2):459–68.PubMedCrossRef
97.
Zurück zum Zitat Suh J, et al. ADAM10 missense mutations potentiate beta-amyloid accumulation by impairing prodomain chaperone function. Neuron. 2013;80(2):385–401.PubMedPubMedCentralCrossRef Suh J, et al. ADAM10 missense mutations potentiate beta-amyloid accumulation by impairing prodomain chaperone function. Neuron. 2013;80(2):385–401.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Slack BE, Ma LK, Seah CC. Constitutive shedding of the amyloid precursor protein ectodomain is up-regulated by tumour necrosis factor-alpha converting enzyme. Biochem J. 2001;357(Pt 3):787–94.PubMedPubMedCentralCrossRef Slack BE, Ma LK, Seah CC. Constitutive shedding of the amyloid precursor protein ectodomain is up-regulated by tumour necrosis factor-alpha converting enzyme. Biochem J. 2001;357(Pt 3):787–94.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Allinson TM, et al. ADAMs family members as amyloid precursor protein alpha-secretases. J Neurosci Res. 2003;74(3):342–52.PubMedCrossRef Allinson TM, et al. ADAMs family members as amyloid precursor protein alpha-secretases. J Neurosci Res. 2003;74(3):342–52.PubMedCrossRef
100.
Zurück zum Zitat Endres K, et al. Shedding of the amyloid precursor protein-like protein APLP2 by disintegrin-metalloproteinases. FEBS J. 2005;272(22):5808–20.PubMedCrossRef Endres K, et al. Shedding of the amyloid precursor protein-like protein APLP2 by disintegrin-metalloproteinases. FEBS J. 2005;272(22):5808–20.PubMedCrossRef
101.
Zurück zum Zitat Vincent B, Checler F. Alpha-secretase in Alzheimer's disease and beyond: mechanistic, regulation and function in the shedding of membrane proteins. Curr Alzheimer Res. 2012;9(2):140–56.PubMedCrossRef Vincent B, Checler F. Alpha-secretase in Alzheimer's disease and beyond: mechanistic, regulation and function in the shedding of membrane proteins. Curr Alzheimer Res. 2012;9(2):140–56.PubMedCrossRef
102.
Zurück zum Zitat Chesebro B, et al. Fatal transmissible amyloid encephalopathy: a new type of prion disease associated with lack of prion protein membrane anchoring. PLoS Pathog. 2010;6(3):e1000800.PubMedPubMedCentralCrossRef Chesebro B, et al. Fatal transmissible amyloid encephalopathy: a new type of prion disease associated with lack of prion protein membrane anchoring. PLoS Pathog. 2010;6(3):e1000800.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Rangel A, et al. Distinct patterns of spread of prion infection in brains of mice expressing anchorless or anchored forms of prion protein. Acta Neuropathol Commun. 2014;2:8.PubMedPubMedCentralCrossRef Rangel A, et al. Distinct patterns of spread of prion infection in brains of mice expressing anchorless or anchored forms of prion protein. Acta Neuropathol Commun. 2014;2:8.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Harris B, Pereira I, Parkin E. Targeting ADAM10 to lipid rafts in neuroblastoma SH-SY5Y cells impairs amyloidogenic processing of the amyloid precursor protein. Brain Res. 2009;1296:203–15.PubMedCrossRef Harris B, Pereira I, Parkin E. Targeting ADAM10 to lipid rafts in neuroblastoma SH-SY5Y cells impairs amyloidogenic processing of the amyloid precursor protein. Brain Res. 2009;1296:203–15.PubMedCrossRef
106.
Zurück zum Zitat Kojro E, et al. Low cholesterol stimulates the nonamyloidogenic pathway by its effect on the alpha-secretase ADAM 10. Proc Natl Acad Sci U S A. 2001;98(10):5815–20.PubMedPubMedCentralCrossRef Kojro E, et al. Low cholesterol stimulates the nonamyloidogenic pathway by its effect on the alpha-secretase ADAM 10. Proc Natl Acad Sci U S A. 2001;98(10):5815–20.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Seipold L, Saftig P. The emerging role of Tetraspanins in the proteolytic processing of the amyloid precursor protein. Front Mol Neurosci. 2016;9:149.PubMedPubMedCentralCrossRef Seipold L, Saftig P. The emerging role of Tetraspanins in the proteolytic processing of the amyloid precursor protein. Front Mol Neurosci. 2016;9:149.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Matthews AL, et al. Regulation of a disintegrin and metalloproteinase (ADAM) family sheddases ADAM10 and ADAM17: the emerging role of tetraspanins and rhomboids. Platelets. 2016:1–9. Matthews AL, et al. Regulation of a disintegrin and metalloproteinase (ADAM) family sheddases ADAM10 and ADAM17: the emerging role of tetraspanins and rhomboids. Platelets. 2016:1–9.
109.
Zurück zum Zitat Saint-Pol J, et al. Regulation of the trafficking and the function of the metalloprotease ADAM10 by tetraspanins. Biochem Soc Trans. 2017;45(4):937–44.PubMedCrossRef Saint-Pol J, et al. Regulation of the trafficking and the function of the metalloprotease ADAM10 by tetraspanins. Biochem Soc Trans. 2017;45(4):937–44.PubMedCrossRef
110.
Zurück zum Zitat Morris RJ, Parkyn CJ, Jen A. Traffic of prion protein between different compartments on the neuronal surface, and the propagation of prion disease. FEBS Lett. 2006;580(23):5565–71.PubMedCrossRef Morris RJ, Parkyn CJ, Jen A. Traffic of prion protein between different compartments on the neuronal surface, and the propagation of prion disease. FEBS Lett. 2006;580(23):5565–71.PubMedCrossRef
111.
Zurück zum Zitat Taylor DR, Hooper NM. The low-density lipoprotein receptor-related protein 1 (LRP1) mediates the endocytosis of the cellular prion protein. Biochem J. 2007;402(1):17–23.PubMedPubMedCentralCrossRef Taylor DR, Hooper NM. The low-density lipoprotein receptor-related protein 1 (LRP1) mediates the endocytosis of the cellular prion protein. Biochem J. 2007;402(1):17–23.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Beland M, Roucou X. The prion protein unstructured N-terminal region is a broad-spectrum molecular sensor with diverse and contrasting potential functions. J Neurochem. 2012;120(6):853–68.PubMed Beland M, Roucou X. The prion protein unstructured N-terminal region is a broad-spectrum molecular sensor with diverse and contrasting potential functions. J Neurochem. 2012;120(6):853–68.PubMed
113.
Zurück zum Zitat Dias MV, et al. PRNP/prion protein regulates the secretion of exosomes modulating CAV1/caveolin-1-suppressed autophagy. Autophagy. 2016;12(11):2113–28.PubMedPubMedCentralCrossRef Dias MV, et al. PRNP/prion protein regulates the secretion of exosomes modulating CAV1/caveolin-1-suppressed autophagy. Autophagy. 2016;12(11):2113–28.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Wetzel S, Seipold L, Saftig P. The metalloproteinase ADAM10: a useful therapeutic target? Biochim Biophys Acta. 2017; Wetzel S, Seipold L, Saftig P. The metalloproteinase ADAM10: a useful therapeutic target? Biochim Biophys Acta. 2017;
115.
Zurück zum Zitat Bove-Fenderson E, et al. Cellular prion protein targets amyloid-beta fibril ends via its C-terminal domain to prevent elongation. J Biol Chem. 2017; Bove-Fenderson E, et al. Cellular prion protein targets amyloid-beta fibril ends via its C-terminal domain to prevent elongation. J Biol Chem. 2017;
116.
Zurück zum Zitat Wang J, et al. Treatment of SMB-S15 cells with resveratrol efficiently removes the PrP(Sc) accumulation in vitro and prion infectivity in vivo. Mol Neurobiol. 2016;53(8):5367–76.PubMedCrossRef Wang J, et al. Treatment of SMB-S15 cells with resveratrol efficiently removes the PrP(Sc) accumulation in vitro and prion infectivity in vivo. Mol Neurobiol. 2016;53(8):5367–76.PubMedCrossRef
117.
Zurück zum Zitat Muller UC, Deller T, Korte M. Not just amyloid: physiological functions of the amyloid precursor protein family. Nat Rev Neurosci. 2017;18(5):281–98.PubMedCrossRef Muller UC, Deller T, Korte M. Not just amyloid: physiological functions of the amyloid precursor protein family. Nat Rev Neurosci. 2017;18(5):281–98.PubMedCrossRef
118.
Zurück zum Zitat Crawford HC, et al. ADAM10 as a therapeutic target for cancer and inflammation. Curr Pharm Des. 2009;15(20):2288–99.PubMedCrossRef Crawford HC, et al. ADAM10 as a therapeutic target for cancer and inflammation. Curr Pharm Des. 2009;15(20):2288–99.PubMedCrossRef
119.
Zurück zum Zitat Atapattu L, et al. An activated form of ADAM10 is tumor selective and regulates cancer stem-like cells and tumor growth. J Exp Med. 2016;213(9):1741–57.PubMedPubMedCentralCrossRef Atapattu L, et al. An activated form of ADAM10 is tumor selective and regulates cancer stem-like cells and tumor growth. J Exp Med. 2016;213(9):1741–57.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Wu GR, et al. Prion protein is required for tumor necrosis factor alpha (TNFalpha)-triggered nuclear factor kappa B (NF-kappaB) signaling and cytokine production. J Biol Chem. 2017; Wu GR, et al. Prion protein is required for tumor necrosis factor alpha (TNFalpha)-triggered nuclear factor kappa B (NF-kappaB) signaling and cytokine production. J Biol Chem. 2017;
124.
Zurück zum Zitat Yang X, et al. Prion protein family contributes to tumorigenesis via multiple pathways. Adv Exp Med Biol. 2017;1018:207–24.PubMedCrossRef Yang X, et al. Prion protein family contributes to tumorigenesis via multiple pathways. Adv Exp Med Biol. 2017;1018:207–24.PubMedCrossRef
Metadaten
Titel
Structural and mechanistic aspects influencing the ADAM10-mediated shedding of the prion protein
verfasst von
Luise Linsenmeier
Behnam Mohammadi
Sebastian Wetzel
Berta Puig
Walker S. Jackson
Alexander Hartmann
Keiji Uchiyama
Suehiro Sakaguchi
Kristina Endres
Jörg Tatzelt
Paul Saftig
Markus Glatzel
Hermann C. Altmeppen
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Neurodegeneration / Ausgabe 1/2018
Elektronische ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-018-0248-6

Weitere Artikel der Ausgabe 1/2018

Molecular Neurodegeneration 1/2018 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Wartezeit nicht kürzer, aber Arbeit flexibler

Psychotherapie Medizin aktuell

Fünf Jahren nach der Neugestaltung der Psychotherapie-Richtlinie wurden jetzt die Effekte der vorgenommenen Änderungen ausgewertet. Das Hauptziel der Novellierung war eine kürzere Wartezeit auf Therapieplätze. Dieses Ziel wurde nicht erreicht, es gab jedoch positive Auswirkungen auf andere Bereiche.

„Restriktion auf vier Wochen Therapie bei Schlaflosigkeit ist absurd!“

06.05.2024 Insomnie Nachrichten

Chronische Insomnie als eigenständiges Krankheitsbild ernst nehmen und adäquat nach dem aktuellen Forschungsstand behandeln: Das forderte der Schlafmediziner Dr. Dieter Kunz von der Berliner Charité beim Praxis Update.

Stuhltransfusion könnte Fortschreiten von Parkinson-Symptomen bremsen

03.05.2024 Parkinson-Krankheit Nachrichten

Kann eine frühzeitige Stuhltransplantation das Fortschreiten von Parkinson-Symptomen verlangsamen? Die Ergebnisse einer randomisierten Phase-2-Studie scheinen dafür zu sprechen.

Frühe Tranexamsäure-Therapie nützt wenig bei Hirnblutungen

02.05.2024 Hirnblutung Nachrichten

Erhalten Personen mit einer spontanen Hirnblutung innerhalb von zwei Stunden nach Symptombeginn eine Tranexamsäure-Therapie, kann dies weder die Hämatomexpansion eindämmen noch die Mortalität senken.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.