• No results found

VU Research Portal

N/A
N/A
Protected

Academic year: 2021

Share "VU Research Portal"

Copied!
27
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

VU Research Portal

Novel Biochemical Signatures of Early Stages of Alzheimer's Disease

Del Campo Milan, M.

2015

document version

Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)

Del Campo Milan, M. (2015). Novel Biochemical Signatures of Early Stages of Alzheimer's Disease.

General rights

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain

• You may freely distribute the URL identifying the publication in the public portal ?

Take down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address:

(2)

Chapter 6

SPPL2b is drastically increased in early stages

of Alzheimer’s disease and associated

with tau pathology.

Under review

(3)

Chapter 6

Abstract

Alzheimer’s disease (AD) is the most common form of dementia but the etiological factors underlying its neuropathology remain unknown. SPPL2b is a novel intramembrane protease targeting tumor necrosis factor alpha and BRI2, both involved in early stages of AD. Here, we investigated the possible role of SPPL2b in AD pathogenesis. Extensive immunohistochemical characterization of SPPL2b showed an increase of up to 10-fold in AD hippocampus (n = 12; p < 0.0001) compared to controls (n = 15). The SPPL2b increase started in very early pathological stages (Braak II-III) and correlated with both neurofibrillary tangles (NFT, p < 0.0001) and amyloid-β (Aβ) plaque formation (p < 0.0001). SPPL2b was also expressed, but at a much lower extent, in other protein-misfolded dementias, such as different types of frontotemporal lobar degeneration and Parkinson’s disease (n = 2/ group), where it was associated with the characteristic protein aggregates. Colocalization and immunoprecipitation studies indicate that SPPL2b is closely associated with tau pathology. In human cerebrospinal fluid (CSF), SPPL2b was significantly decreased in AD (n = 14) compared to controls (n = 10), and correlated with both Aβ42 CSF concentration (r = 0.60, p = 0.005) and MMSE scores (r = 0.47, p = 0.02). In conclusion, our data revealed drastic changes in SPPL2b associated with both the development of protein aggregates and cognitive impairment in early stages of AD, suggesting a novel, important role of SPPL2b not only in AD etiology, but also in the development of other protein-misfolded dementias.

(4)

Introduction

Alzheimer’s disease (AD) is an age-related irreversible neurodegenerative dementia characterized by the accumulation and aggregation of amyloid β peptide (Aβ) in senile plaques and hyperphosphorylated tau (p-tau) in neurofibrillary tangles (NFTs)1,2.

The etiological factors underlying AD neuropathology remain unknown. Thus, the understanding of AD pathogenesis is a challenge and the quest for novel proteins involved in AD is still ongoing. We recently observed that SPPL2b levels were increased as much as 10-fold in AD hippocampus compared to controls3. SPPL2b belongs to the

intramembrane-cleaving GXGD-aspartyl protease family, which also includes its homologues SPPL2a, c and SPPL3, the signal peptide peptidase (SPP) and the Alzheimer’s disease-associated presenilins (PS1 and PS2)4–8. These enzymes are involved in the regulated intramembrane

proteolysis

RIP) of transmembrane proteins. RIP generally starts with an initial shedding of the ectodomain followed by intramembrane proteolysis that liberates the intracellular domain (ICD) and a small peptide corresponding to the transmembrane region between the two cleavage sites9. RIP is an essential mechanism for several and different physiological

pathways such as lipid metabolism10, cell cycle11, inflammation12,13 or cell differentiation14

and dysfunctions in RIP have already been linked to different disorders including AD, where RIP of APP is a classical example15.

Despite the recently developed cellular and mice models, the biological function of the SPPL proteins remains largely unknown7. SPPL2a and SPPL2b share two substrates related

to AD: the tumor necrosis factor α (TNFα)12,13,16 and BRI217. While SPPL2a is ubiquitously

expressed in different murine tissues with the lowest expression found in the brain, SPPL2b is mainly expressed in brain tissue. Importantly, SPPL2b expression was especially pronounced in the murine hippocampus18, suggesting a role of SPPL2b, rather than

SPPL2a, in learning and memory.

The first SPPL2b substrate, TNFα, is upregulated in early AD16, and it participates in

different pathways relevant for AD pathogenesis including inflammation19,20, regulation of

the Aβ precursor protein (APP) processing21 and the aggregation of tau22. TNFα is initially

processed by ADAM17 leading to the release of the TNFα soluble form23. SPPL2b cleavage

induces the release of TNFα-ICD, which promotes the expression of the pro-inflammatory cytokine interleukin-12 (IL-12) essential for the adaptive immunity12. Interestingly,

the IL-12 pathway was able to regulate not only Aβ load but also cognitive function in mice model of AD and aging20,24. Taken together, changes in TNFα processing may have

(5)

Chapter 6

The other SPPL2b substrate, BRI2, can also regulate the homeostasis of critical proteins involved in AD pathogenesis, i.e. APP processing25–31, Aβ formation and fibrillation32–34,

insulin degrading enzyme secretion35 or the expression of β-secretase 136. Moreover, the

overexpression of BRI2 in AD mouse models reduced AD pathology27,32,35, and mutations

in BRI2 coding genes lead to the development of familial British and Danish dementias (FBD and FDD)37,38. Interestingly, memory deficits and impaired synaptic plasticity in

FBD and FDD mice models were caused by loss of wild-type BRI2 function rather than amyloidogenesis of the mutated fragments, suggesting that BRI2 has a key role in memory performance39,40. Importantly, we recently found aggregates of BRI2 ectodomain

in Aβ plaques in early stages of AD. We proposed that the observed decrease in the levels of the enzymes involved in the shedding of BRI2 ectodomain (furin and ADAM103,17,41–43)

together with the drastic increase in the levels of SPPL2b3, may enhance the release of an

un-processed BRI2 ectodomain instead of the shorter BRI2 peptides in AD. The larger BRI2 fragment may then aggregate leading to the observed BRI2 deposits and a loss of BRI2 function as shown by the loss of BRI2-APP complexes in AD3.

Thus, previous studies indicate that SPPL2b substrates can play important, independent roles in the development of AD and we recently observed a dramatic increase in SPPL2b in AD tissue. Since a link between SPPL2b and AD has not yet been anticipated, we performed an extensive characterization of SPPL2b in human brain tissue and cerebrospinal fluid (CSF) from controls and AD patients at different stages of the disease. We also investigated the relation of SPPL2b with tau, a classical protein involved not only in AD, but also in other protein-misfolded dementias.

Materials and Methods

Post-mortem brain tissue

Post-mortem brain material was obtained from the Netherlands Brain Bank (Amsterdam, The Netherlands). All donors (n = 42) or their next of kin provided written informed consent for brain autopsy and use of tissue and medical records for research purposes. Clinical diagnosis and neuropathological evaluation was performed on formalin-fixed, paraffin-embedded tissue from different brain areas as previously described3. Brain tissue

(6)

Table 1. Demographic data of post-mortem tissue

Patient Clinical Pathological

Gender Age PMI(h) Grade Grade

number diagnosis diagnosis Brain area (Braak, NFT) (Thal, Aβ)

1 CON CON M 96 6,3 Hipp I 0

2 CON CON M 81 5,3 Hipp II 0

3 CON CON M 56 10 Hipp 0 0

4 CON CON M 56 9,15 Hipp 0 4

5 CON CON M 66 7,45 Hipp 0 1

6 CON CON F 89 6,25 Hipp II 3

7 CON CON F 94 4,05 Hipp I 0

8 CON CON F 89 3,52 Hipp III 0

9 CON CON M 88 4,43 Hipp II 1

10 CON CON F 77 2,55 Hipp I 1

11 CON CON F 84 6,55 Hipp I 0

12 CON CON M 88 7 Hipp III 1

13 CON CON F 73 7,45 Hipp I 3

14 CON Medium F 86 6,25 Hipp III 1

15 CON CON M 74 5 Hipp,Occ,Par III 3

16 CON CON F 93 5,5 Hipp,Occ,Temp II 0

17 CON CON F 77 na Occ,Par I 1

18 AD AD F 68 3,5 Hipp VI 4 19 AD AD F 67 5,5 Hipp VI 4 20 AD AD F 78 4,5 Hipp V 4 21 AD AD M 93 4,3 Hipp V 4 22 AD AD F 93 2,3 Hipp IV 4 23 AD AD F 91 5,45 Hipp VI 4 24 AD AD F 86 5,55 Hipp IV 4 25 AD AD M 61 4 Hipp V 4 26 AD AD M 74 5,35 Hipp VI 4 27 AD AD F 72 5,55 Hipp VI 4 28 VD AD F 92 4,15 Hipp IV 4 29 VD AD F 91 6,05 Hipp IV 4 30 AD AD M 59 5,05 Temp, Par VI 4 31 AD AD F 75 39 Temp, Par VI 4 32 AD AD F 62 5,55 Temp, Par VI 4 33 AD AD F 89 4,3 Occ,Par VI 4 34 AD AD F 86 5,05 Temp, Occ IV 4 35 PD PD M 62 <24 Mes III na 36 PD PD M 55 Mes II na 37 FTD FTLD-tau F 66 6,4 Front na na 38 FTD FTLD-tau M 46 5,35 Front na na

39 PiD FTLD-tau M 70 5,15 Front na na

40 PiD FTLD-tau M 82 4,1 Front II na

41 PSP PSP M 75 5,05 Put I na

42 PSP PSP M 80 4,5 Put I na

(7)

Chapter 6

Human CSF samples

CSF material was obtained from Amsterdam Dementia Cohort44, NUBIN/ (NeuroUnit

Biomarkers for Inflammation and Neurodegeneration) VUmc biobank (Amsterdam, The Netherlands). Clinical assessment of subjects and collection, storage and analysis of CSF biomarkers (Aβ42, t-Tau and p-Tau) were done as previously described45. CSF samples were

stored in agreement with JPND-BIOMARKAPD guidelines46. Cognitively normal subjects

(subjective memory complaints (SMC), n = 10) and AD patients (n = 14) were selected. Diagnoses were defined in a multidisciplinary meeting as described before44. Age, gender,

biomarker levels as well as Mini Mental State Examination (MMSE) scores of all cases are listed in Table 2. The ethical review board of the VUmc approved the study, and all subjects gave written informed consent.

Table 2. Demographic data of CSF samples

Patient

Groups (mean ± SD) No(M/F)Age (mean± SD)MMSE (pg/mL)Aβ42 (pg/mL)t-Tau (pg/ml)p-Tau

SMC 58 .1± 1.15 10(7/3) 27.5 ± 1.27 915.4± 221.7 208.1± 74.62 44.22± 19.8 AD 57.1 ± 1.36 14(8/6) 18.79± 3.12a 481.5± 81.84a 862.2± 409.8a 97.18± 33.5a

Data are reported as means ± standard deviation (SD). SMC = Subjective memory complaints, AD = Alzheimer’s disease. MMSE = Mini Mental Score Examination. a = at least p < 0.05 from SMC.

Western blotting.

Preparation of human hippocampus tissue (12 µg) for western blotting was performed as previously described3. CSF samples (45 µl) were prepared in sample buffer (2% SDS, 0.03

M Tris, 5% 2-Mercaptoethanol, 10% glycerol, bromophenol blue) and heated 15 min at 50ºC. Electrophoresis was carried out in 10% SDS-PAGE mini gels and immunodetection was performed using LI-COR Odyssey system (LI-COR bioscience, Lincoln, Nebraska USA) following the manufacturer’s instructions. The following primary antibodies were used: rabbit anti-human SPPL2b N-terminal (1 µg/ml; ARP44989, epitope detected between amino acids 106-155; Aviva systems biology, San Diego, CA, USA), mouse anti-SPPL2a199-217 (1:50;47) and rabbit anti-human SPPL2a

504-520 (1:500;18). Specificity of the anti-SPPL2b

antibody on Western blot was analyzed through reactivity comparison with mouse and rabbit anti-SPPL2a against human hippocampus, the closets homologue to SPPL2b6.

(8)

Immunohistochemistry

Formalin-fixed and paraffin-embedded hippocampus, parietal, occipital and temporal cortex sections (5µm) were mounted on Superfrost plus tissue slides (Menzel-Glaser, Braunschweig, Germany) and dried overnight at 37°C. Single and double immunohistochemistry procedures are described in detail in supporting information. The following primary antibodies were used: rabbit anti-SPPL2b N-terminal (0.5 µg/ml; Aviva systems biology) and mouse anti-AT8 antibody (1:400, Thermo Scientific). Antibody pre-absorption with 1, 10, 100 and 200 molar excess of SPPL2b antigenic peptide (Aviva Systems biology) was also performed for the anti-SPPL2b antibody.

Evaluation of stainings

Quantitative analysis of SPPL2b immunoreactivity was performed on single stained slides by manually counting the number of SPPL2b positive structures in different regions of the hippocampus (CA4-CA2, CA1 and Subiculum), correcting for the size of the area. Contiguous microscopic fields were evaluated using a 10x objective (0.64 mm2). The

researcher performing either the staining or the counting was blinded for the diagnoses and other characteristics of the cases. Double immunohistochemistry was analyzed using Nuance Fx spectral imaging camera (Perkin Elmer, Waltham, MA, USA) on a DM5000B microscope (Leica, Wetzlar, Germany).

SPPL2b immunoprecipitation.

(9)

Chapter 6

Statistical analysis.

Statistical analyses were performed on SPSS version 20.0 (Chicago, USA) using non-parametric Mann-Whitney U (two groups analysis) or Kruskall-Wallis (multiple group analysis) tests to analyze group differences. Correlation analyses were done using Spearman’s test. CSF data were normally distributed and thus, parametric t-tests or Pearson’s correlations was used. Values with p < 0.05 were considered significant.

Results

Antibody characterization

Analysis of the amino acidic sequence  of the antigenic SPPL2b peptide using Basic Local Alignment Search Tool (BLAST, NCBI) showed an e-value of 1e-26 for SPPL2b. No other proteins were identified, indicating that the antigenic synthetic peptide used for antibody production is a unique sequence for SPPL2b. Specificity of the anti-SPPL2b antibody was confirmed by a complete abolishment of signal after pre-absorption with the antigenic peptide in both western blot and immunohistochemistry analyses (Fig. 1a,b). The immunoreactivity observed using two different anti-SPPL2a antibodies was completely different from the one observed with anti-SPPL2b antibody in Western blot and immunohistochemistry (Fig. 1c-e). These data indicate that the antibody used is specific for SPPL2b and does not cross-react with its closest family homologue SPPL2a. SPPL2b expression is increased in early stages of AD and correlates with BRI2 deposition

In order to further understand and characterize the increase in SPPL2b expression in AD tissue3, we performed SPPL2b immunostainings on post-mortem hippocampus brain

sections from twenty seven patients, which were all previously used in our Western blot study3. Three different areas within the hippocampus (CA4 to 2, CA1 and Subiculum) were

(10)

cortex. The SPPL2b expression in these areas was considerably lower (approximately 90% less) compared to that observed in the hippocampal areas and mainly showed a globular-like staining pattern (Fig. 2e). No SPPL2b staining was observed in those areas when control cases were analyzed (data not shown).

Semi-quantitative analysis confirmed the significant increase of SPPL2b expression in AD compared to control patients in all hippocampal areas when patients were grouped either by the pathological (Fig. 3a) or clinical diagnosis (data not shown). There was a significant correlation between the reactivity of SPPL2b staining and the levels of SPPL2b expression observed by Western blot (r = 0.82, p < 0.0001, data not shown).

We next questioned in which stage of the disease SPPL2b immunoreactivity becomes detectable. Hippocampal SPPL2b expression was already increased in very early stages of AD (II/III-3) and correlated with both tangle and amyloid pathology (Fig 3b, c). Related to NFT formation, SPPL2b was already increased in very early stages (Braak II-III), and reached its maximum at Braak stages IV and V. Additionally, a drastic increase in SPPL2b expression was observed between Braak III and IV (Fig. 3c). Moreover, we observed a strong correlation (r = 0.813, p<0.0001) between the immunoreactivity of SPPL2b and BRI2 deposition (Fig. 3d) in the same patients3. Collectively, these results revealed that increased hippocampal

1 250 - 150 - 100 - 75 - 25 - 50 - 37 - 2 SPPL2b A B 1 2 Preabs SPPL2b SPPL2b Pre-absorbed SPPL2b AD Cn AD Cn SPPL2b Ms AD Cn SPPL2a Rb 250 - 150 - 100 - 75 - 50 - 37 - SPPL2b SPPL2a C D Fig. 1

Fig. 1 SPPL2b antibody characterization

A-B, Post-mortem hippocampus from AD patients were analyzed by western blot (A) or immunohistochemistry

(11)

Chapter 6 A   Con tr ol

 

( SP PL2b )   AD  ( SP PL2 b )   Br aak  s tag es  

+  

-­‐  

0-­‐0   III-­‐ 0   IV -­‐C   V-­‐ C  

CA4-­‐2   CA1   Subiculum  

Parietal   Temporal  

B   C   D   E  

(12)

SPPL2b expression, which strongly correlated with the presence of BRI2 deposits, starts in very early stages of AD.

SPPL2b expression in other protein-misfolded dementias

We next investigated whether SPPL2b expression was specific for AD pathology or if it was also present in the most affected areas of other protein-misfolding neurodegenerative dementias, such as FTD, PiD, PSP and PD (n=2/group). Interestingly, a noticeable, though clearly less prominent, SPPL2b staining was also observed in all the cases analyzed (Fig. Fig. 2 Different SPPL2b histological lesions in post-mortem AD hippocampus

A, SPPL2b expression in post-mortem hippocampus sections from control (Braak 0 and III; upper rows) and AD

cases (Braak IV and V; bottom rows). Three different hippocampal areas were analyzed: CA4-2, CA1 and Subiculum.

SPPL2b staining was present in all AD hippocampal areas, but not in controls. B-D, Higher magnification shows different SPPL2b histological lesions showing a (B) globular like-, (C) tangle like- and (D) neuritic plaque like- pattern. E, SPPL2b staining in the temporal and parietal cortex from an AD patient. Squares within the sections represent a higher magnification of the observed SPPL2b staining. Scale bars: a-e, 100µm; b-e, 0.1 µm.

Fig.  3   B   0 I II III IV V VI 0 20 40 60 100 150 200 250 p < 0.0001 Braak stages (NFT) SPPL 2 b I R (A rb it rary u n it s) * * ** C   D   A   Control AD 0 10 20 30 40 CA4 SPPL 2 I R (A rb it rary u n it s) **** Control AD 0 20 40 60 80 100 CA1 *** Control AD 0 50 100 150 Subiculum *** 0 1 3 4 0 50 100 150 200 250

Thal staging (amyloid)

SPPL 2 b I R (A rb it rary u n it s) *** ** p    <  0,0001     0 50 100 150 200 250 0 10 20 30 40 SPPL2b IR (Arbitrary units) BRI 2 I R (A rb it rary u n it s) r  =    0,813   p    <  0,0001     Fig.  3   B   0 I II III IV V VI 0 20 40 60 100 150 200 250 p < 0.0001 Braak stages (NFT) SPPL 2 b I R (A rb it rary u n it s) * * ** C   D   A   Control AD 0 10 20 30 40 CA4 SPPL 2 I R (A rb it rary u n it s) **** Control AD 0 20 40 60 80 100 CA1 *** Control AD 0 50 100 150 Subiculum *** 0 1 3 4 0 50 100 150 200 250

Thal staging (amyloid)

SPPL 2 b I R (A rb it rary u n it s) *** ** p    <  0,0001     0 50 100 150 200 250 0 10 20 30 40 SPPL2b IR (Arbitrary units) BRI 2 I R (A rb it rary u n it s) r  =    0,813   p    <  0,0001     Fig.  3   B   0 I II III IV V VI 0 20 40 60 100 150 200 250 p < 0.0001 Braak stages (NFT) SPPL 2 b I R (A rb it rary u n it s) * * ** C   D   A   Control AD 0 10 20 30 40 CA4 SPPL 2 I R (A rb it rary u n it s) **** Control AD 0 20 40 60 80 100 CA1 *** Control AD 0 50 100 150 Subiculum *** 0 1 3 4 0 50 100 150 200 250

Thal staging (amyloid)

SPPL 2 b I R (A rb it rary u n it s) *** ** p    <  0,0001     0 50 100 150 200 250 0 10 20 30 40 SPPL2b IR (Arbitrary units) BRI 2 I R (A rb it rary u n it s) r  =    0,813   p    <  0,0001    

Fig. 3 SPPL2b expression is increased in early stages of AD and correlated with BRI2 deposition

A, SPPL2b immunoreactivity (IR) was semiquantitatively measured for each patient in each hippocampus area,

(13)

Chapter 6

4a). SPPL2b staining patterns seemed to be associated with the main pathological characteristics of these disorders: aggregated tau in FTD, pick’s bodies in PiD, coiled bodies and tau inclusions in glia cells in PSP and Lewy bodies and α-synuclein inclusions in PD48,49 (Fig. 4b). Taken together, the results revealed that a lower but noticeable SPPL2b

expression was also observed in other protein-misfolding neurodegenerative dementias, which appears associated with the characteristic protein aggregates of those disorders. SPPL2b binds tau and is associated with hyperphosphorylated tau in AD and other tauopathies

Since the SPPL2b staining patterns in various dementias resembled the pattern observed for tau pathology, we further investigated whether SPPL2b was associated with tau. SPPL2b was successfully immunoprecipitated from post-mortem human hippocampus (Fig. 5a). Immunoprecipitation of SPPL2b revealed that SPPL2b binds tau and that tau-SPPL2b complexes were principally found in AD tissue compared to controls (Fig. 5b). The lack of signal in the SPPL2b IP samples of other transmembrane proteins such as APP together with the recovery of the signal using an anti p-Tau antibody further supported the specificity of the binding between SPPL2b and tau and further extends the binding to the p-Tau form (Figure 5c). Double immunohistochemistry experiments showed a clear co-localization between SPPL2b expression and p-Tau in NFTs in the different brain areas of AD patients (Fig. 5d). Moreover, co-localization of SPPL2b with tau aggregates was also observed in the different tauopathies (Fig. 5e). No staining was observed in the same area in control patients (data not shown). These data revealed that SPPL2b is a novel tau-binding protein that associates with p-Tau in pathological conditions.

SPPL2b expression is decreased in CSF and correlates with Aβ42 and cognitive decline. Since SPPL2b is a multi-pass transmembrane protein, its detection in CSF is challenging. However, integral membrane proteins such as PS1 have been previously detected in CSF by western blot using a specific pre-treatment of CSF (i.e. heating samples at 50ºC during 15 min)50. We applied this pre-treatment to detect SPPL2b in CSF. Western blot analyses

(14)

FTD

PD

PSP

PiD

AD

AD

A PSP PiD PD PD B FTD PSP

Fig. 4

Fig. 4 Different SPPL2b histological lesions are also observed in other protein-misfolded dementias A, SPPL2b expression was also observed in post-mortem brain tissue from patients with FTD (frontal lobe), PiD

(15)

Chapter 6 Fig. 5 1 Cn 2 1 AD 2 1 2 1 AD 2 Cn Untreated SPPL2b IP t-Tau 3 Cn 4 3 AD 4 3Cn 4 3 AD 4 t-Tau A AT8 SPPL2b SPPL2b AT8 D Pi D PSP FTD AT8 SPPL2b SPPL2b AT8 E 3 Cn 4 3 AD 4 AD4 3 4 Cn 3 4 AD Untreated SPPL2b IP p-Tau APP 3 1 P1 Cn P2 -Ctr l (no ab ) AD 150 - 100 - 75 - 50 - 37 - * SPPL2b SPPL2b IP B C - Ctr l ( Rb -IG) SPPL2b IP Untreated AD2 - Ctr l ( Rb -IG) AD4 - Ctr l

Fig. 5 SPPL2b binds tau in AD and its expression is associated with hyperphosphorylated tau in AD and other tauopathies.

A, SPPL2b was immunoprecipitated from human hippocampus of 2 individual control cases (Cn 3 and 1) and

2 different pools of AD cases (P1 and P2) using anti-SPPL2b antibody. Negative control was performed using beads without antibody B, Total tau (phosphorylated and non phosphorylated) was analyzed in post-mortem hippocampus form 4 individual control cases (Cn 1-4) and 4 AD patients (AD 1-4) by Western blot in the original samples (untreated) and in the immunoprecipitated-SPPL2b samples (SPPL2b IP). C, APP (upper) and p-Tau (bottom) were analyzed in post-mortem hippocampus form 2 individual control cases (Cn 3 and 4) and 2 AD patients (AD 3 and 4) by Western blot in the original samples (untreated) and in the immunoprecipitated-SPPL2b samples (immunoprecipitated-SPPL2b IP). Negative controls (- Ctr, Rb-Ig) are IPs performed with an irrelevant rabbit antibody using an AD sample. D-E, Double immunohistochemistry and spectral imager analysis showed that SPPL2b expression was associated with phosphorylated tau (AT8) in AD (D) and in the different tauopathies analyzed (E). *: Unspecific band. Scale bars: a, 10µm; b, 100 µm.

Fig.  6   SMC AD AD SMC 50 - 100 - 150 - A! SMC AD 0 1 2 3 S P P L2b 60 k Da (A rb it ra ry u n it s) * 0 500 1000 1500 0 1 2 3 r = 0.603 p = 0.005 Aβ42(pg/ml) S P P L2b 60k Da (a rb it ra ry u n it s) C! B! D! 10 15 20 25 30 35 0 1 2 3 r = 0.47 p = 0.02 MMSE score S P P L2b 60k Da (a rb it ra ry u n it s)

Fig. 6 SPPL2b is decreased in AD CSF and correlated with Aβ42 CSF concentration and cognitive decline A, Representative Western blot of SPPL2b in CSF from 4 SMC and 3 AD cases indicates three different SPPL2b

isoforms at 60, 100 and 150 kDa. B, Analysis of the 60kDa SPPL2b reactivity in human CSF showed that SPPL2b was significantly decreased in AD (n = 13) compared to control (n = 10) cases. Correlation analysis showed

a significant positive correlation of SPPL2b 60 kDa with Aβ42 CSF concentration (C) and MMSE scores (D).

(16)

Discussion

The quest for additional relevant proteins and pathways involved in the etiology and development of AD is still ongoing. In this study, we unraveled a potentially important role of SPPL2b in AD pathogenesis and protein aggregation, based on the fact that the levels of SPPL2b were drastically increased in human AD hippocampus compared to controls and that the SPPL2b increase started in very early stages of human AD pathology (Braak II-III for NFT). Moreover, in CSF, SPPL2b levels were decreased in AD patients and correlated with the concentration of Aβ42 and cognitive decline. Interestingly, SPPL2b was also present in FTD, PSP, PiD and PD, although the staining was far less intense than in AD. In addition, the data revealed that SPPL2b is a novel tau-binding protein, which is associated with protein aggregates in the different disorders analyzed.

Antibody characterization of the commercially available anti-SPPL2b antibody used in this study showed a different reactivity from that observed with the antibodies against the SPPL2b homologue SPPL2a6 on both Western blot and immunohistochemistry.

Although the expected molecular weight of SPPL2b is 60 kDa, SPPL2b reactivity showed a smear-like reactivity in AD samples which is likely attributed to the high glycosylation pattern of the protein as previously described in cell culture models51. Along with the

almost complete absence of signal following pre-absorbing the SPPL2b antibody with its antigenic peptide, these data support the specificity of this SPPL2b antibody.

In our recent investigations of BRI2, a protein inhibiting Aβ formation25,26, we observed

a 10-fold increase in SPPL2b expression in AD hippocampus homogenates compared to non-demented controls3. In the current study, we performed an extensive

immunohistochemical analysis of SPPL2b and showed a strong SPPL2b immunoreactivity in AD patients in all the hippocampal areas. The reduced SPPL2b staining in the cortex area, where an extensive plaque and NFT pathology can be found52,53, discard the

possibility that the SPPL2b antibody recognize conformational aggregated structures. It is important to note that unlike Aβ deposition54, SPPL2b staining was nearly absent in the

hippocampus of control cases, highlighting the importance that this protein may have not only in the development of AD, but also as a potential marker of AD pathology. We observed that increased SPPL2b levels were detectable at early stages of AD coinciding with the appearance of NFTs in the hippocampus (Braak II and III for NFT53). Moreover, a

drastic SPPL2b increase was also found inbetween Braak stages III and IV, the latter related to the clinical manifestation of dementia55,56. These results, together with the predominant

(17)

Chapter 6

The strong correlation between SPPL2b staining and BRI2 deposits further supports our hypothesis that modification of SPPL2b may affect at least in part the behavior of SPPL2b’s substrates, which ultimately may lead to a dysfunction of the different substrates3.

The two SPPL2b substrates (TNFα and BRI2) are also modified in early stages of AD3,57

and can regulate key molecular processes of AD pathogenesis (i.e. APP processing, Aβ aggregation, NFT formation, inflammation)13,21,22,58. Specifically, the TNFα-ICD released

after SPPL2b cleavage likely plays an important role in AD, as it stimulates the secretion of IL-12, a cytokine pathway regulating the load of Aβ as well as cognitive performance in mice models of AD (APPPS1) and aging (SAMP8)12,20,24 . Whether the BRI2-ICD released after

SPPL2b processing also plays a role in AD has yet to be elucidated since its physiological function remains unknown7. In addition, the drastic changes in SPPL2b may alter the

RIP of BRI2 and thus, its functionality. Reduced BRI2 function could promote not only Aβ production25,26 and fibrillation34 but also memory impairment39,40. Taken together, we

suggest that modifications in the levels of SPPL2b may have major consequences in the diverse functions of TNFα and BRI2, leading to the alteration of multiple independent molecular pathways involved in AD.

SPPL2b was also expressed to a lower extent in other protein-misfolded dementias. The expression of SPPL2b with the main pathological characteristics of those disorders, including tau aggregates, Pick’s bodies and oligodendroglial coiled bodies in FTLD types, as well as α-synuclein inclusions and Lewy bodies in PD48, suggests a more general role of

SPPL2b in protein aggregation mechanisms. This is further supported by the intracellular location of SPPL2b in the endosomes/lysosomes59, where it may participate in the

ubiquitin and autophagy-lysosome pathways60. Although these data show that SPPL2b

changes are probably not specific to AD, SPPL2b may play a more relevant role in AD pathogenesis considering the stronger changes in expression in AD compare to other protein-misfolded dementias and its higher physiological expression in the hippocampus. The different SPPL2b histological patterns observed in AD tissue and in different tauopathies, together with the strong correlation with NFT Braak staging, suggested an association between SPPL2b and tau pathology. This was supported by the clear co-localization observed between phosphorylated tau and SPPL2b in the different tauopathies and the immunoprecipitation experiments, which revealed that SPPL2b is a novel tau binding protein. In addition, SPPL2b-tau binding complexes were principally observed in the AD tissue and not in controls. Other tau binding proteins have been previously identified, which either could promote or prevent tau phosphorylation and aggregation61–63. It remains to be elucidated whether the interaction between SPPL2b

and tau in pathological conditions aims to halt tau misfolding (i.e. via autophagy-lysosome pathways64) or alternatively promotes tau aggregation. Strikingly, while no

(18)

immunoprecipitated from the hippocampus of control patients. However, the reactivity of SPPL2b in the immunoprecipitated samples is likely due to the higher amount of sample used for the immunoprecipitation (5x fold).

Our data showed that different forms of SPPL2b are present in human CSF, including a band observed at the expected molecular weight of 60 kDa. The detection of membrane proteins in CSF might be enigmatic but the presence of extracellular vesicles (i.e. exosomes) in CSF have been reported65. Thus, similarly to other membrane protein such

as APP or different solute carriers66, SPPL2b may reach the CSF via exosomes. Although

the specificity of the different bands needs further confirmation, previous animal models have shown that SPPL2b can also run around 100 kDa18. Similarly to SPP or SPPL3, the

formation of SDS-resistant dimers and trimmers or the presence of glycosyl groups may explain the higher molecular weight forms observed67,68. Interestingly, the 60 kDa form

was significantly decreased in AD CSF, showing an inverse relationship to that observed in human AD tissue. Similar inverse expression patterns between brain tissue and CSF have been previously reported for other proteins such as Aβ4269, which is currently seen as

the earliest biomarker to become abnormal in AD70. The correlation between CSF SPPL2b

reactivity with both the Aβ42 concentration and MMSE scores supports the idea that changes in SPPL2b take place in very early stages of AD and are associated with cognitive decline. However, the lack of correlation between SPPL2b CSF and the concentration of t-Tau or p-Tau in CSF remains elusive. Taken together, the CSF results call for the development of more sensitive techniques for detection of SPPL2b in vivo, in either CSF or brain tissue, to permit further investigation of the potential of SPPL2b as a biomarker in different stages of AD.

In summary, this study reveals that SPPL2b is a novel protein associated with tau pathology, which is already increased in very early stages of AD (Braak II-III). The very small overlap that we found in SPPL2b expression between control and AD patients, together with the correlation of SPPL2b expression with cognitive impairment (Braak III-IV and MMSE) discriminate SPPL2b from other different proteins associated with AD hallmarks (i.e. Apolipoprotein D, αβ-crystallin71,72), and suggest that SPPL2b expression might be an

(19)

Chapter 6

Acknowledgment

We acknowledge Dorine Wouters from the Neurochemistry Laboratory of the Clinical Chemistry department at the VUmc for her technical assistance and John Bol from the Anatomy and Neuroscience department at the Medical Faculty of the VU University for his assistance with the spectral imager.

Funding

This work was supported by the Erasmus Mundus Joint Doctorate Program (EMJD 2009-2013, Action 1B, Grant 159302-1-2009-1-NL-ERA, European Neuroscience Campus Network).

Conflict of interest

Dr. Teunissen serves on the advisory board of Fujirebio and Roche, received research consumables from Euroimmun, IBL, Fujirebio, Invitrogen and Mesoscale Discovery. Dr Scheltens serves/has served on the advisory boards of: Genentech, Novartis, Pfizer, Roche, Danone, Nutricia, Jansen AI, Baxter and Lundbeck. He has been a speaker at symposia organised by Lundbeck, Lilly, Merz, Pfizer, Jansen AI, Danone, Novartis, Roche and Genentech. He serves on the editorial board of Alzheimer’s Research & Therapy and Alzheimer’s Disease and Associated Disorders, is a member of the scientific advisory board of the EU Joint Programming Initiative and the French National Plan Alzheimer. The Alzheimer Center receives unrestricted funding from various sources through the VUmc Fonds.  Dr Scheltens receives no personal compensation for the activities mentioned above.

(20)

Supporting information: Materials and Methods

Immunohistochemistry.

Formalin-fixed and paraffin-embedded human brain sections (5µm) were mounted on Superfrost plus tissue slides (Menzel-Glaser, Braunschweig, Germany) and dried overnight at 37°C. For all stainings, sections were deparaffinized and subsequently immersed in 0.3% H2O2 in methanol for 30 minutes to quench endogenous peroxidase activity. Sections were boiled in a microwave in 10 mmol/L pH 6.0 sodium citrate buffer during 10 minutes for antigen retrieval. Sections were incubated overnight at 4°C with rabbit anti-SPPL2b N-terminal (0.5 µg/ml; Aviva systems biology) in normal antibody diluent (Ymmunologic, Duiven, The Netherlands). After washing with PBS, sections were incubated with EnVision solution (anti-mouse/rabbit HRP, undiluted; DAKO, Glostrup, Denmark) for 60 minutes. Sections were washed with PBS and color was developed using 3,3-diaminobenzidine (DAB, 0.1 mg/ml, 0.02% H2O2, 2 minutes; Sigma, St. Louis, MO) as chromogen. Nuclei were stained with hematoxylin and sections were mounted using Aquamount (BDH Laboratories Supplies, Dorset, UK). Antibody specificity was evaluated by comparing immunohistochemistry patterns using rabbit anti-SPPL2a antibody. Antibody pre-absorption with 1, 10, 100 and 200 molar excess of SPPL2b antigenic peptide (Aviva Systems biology) was also performed for the anti-SPPL2b antibody.

To determine co-localization of SPPL2b with neurofibrillary tangles, sections were first incubated with rabbit anti-SPPL2b (0.5 µg/ml) and color was visualized with DAB as described above. Then, sections were washed with PBS and incubated with mouse anti-AT8 antibody (1:400, Thermo Scientific) overnight at 4°C. After washing with PBS, sections were incubated with biotin-conjugated swine anti mouse-F(ab)2 (1:500 dilution) over 60 minutes for the detection of AT8 antibodies. Sections were then intensively washed with MilliQ water and Tris-HCl buffer (0.2 M, pH 8.5) for 5 minutes before visualization of AT8 with liquid permanent red. Negative controls were generated by omission of primary antibodies.

SPPL2b immunoprecipitation.

(21)

Chapter 6

complexes were then incubated with Protein G PLUS-Agarose beads (1:2) overnight at 4ºC. Beads were washed 4 times with PBS-Tween 0.5% (v/v, PBS-T) buffer and re-suspended in sample buffer. SPPL2b precipitates were then analyzed for SPPL2b immunoreactivity by Western blot using rabbit anti-human SPPL2b N-terminal as described above.

To analyze the binding between SPPL2b and tau, individual human hippocampus homogenates (100 µg) from AD (AD 1-4) and control cases (Cn 1-4) were pre-cleared as described above. Then, sample buffer was added to half of the pre-cleared homogenates (untreated) and stored at -20ºC until further analysis. The other half was incubated with 1 µg of either rabbit anti-SPPL2bor control rabbit polyclonal antibody for 1 hour at 4°C. Antibody-bound protein complexes were then processed as described before. SPPL2b precipitates and untreated samples were analyzed for tau immunoreactivity by Western blot using monoclonal mouse anti-tau (1:500, clone TAU-5, Thermo scientific), which recognizes both phosphorylated and non-phosphorylated forms. In order to test the specificity of binding between SPPL2b and tau, we analyzed the reactivity of APP in SPPL2b precipitates, since APP is a transmembrane type I protein and thus it is not expected to bind SPPL2b, which process type II orientated proteins7. To do so anti-tau

(22)

References

1. Selkoe, D. J. Alzheimer ’ s Disease : Genes , Proteins , and Therapy. 81, 741–766 (2001). 2. Grundke-Iqbal, I. et al. Abnormal phosphorylation of the microtubule-associated protein tau

(tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A 83, 4913–7 (1986).

3. Del Campo, M. et al. BRI2-BRICHOS is increased in human amyloid plaques in early stages of Alzheimer’s disease. Neurobiol Aging 35, 1596–604 (2014).

4. Weihofen, A. & Martoglio, B. Intramembrane-cleaving proteases: controlled liberation of proteins and bioactive peptides. Trends Cell Biol 13, 71–8 (2003).

5. Haass, C. & Steiner, H. Alzheimer disease gamma-secretase: a complex story of GxGD-type presenilin proteases. Trends Cell Biol 12, 556–62 (2002).

6. Martoglio, B. & Golde, T. E. Intramembrane-cleaving aspartic proteases and disease: presenilins, signal peptide peptidase and their homologs. Hum Mol Genet 12 Spec No, R201–6 (2003). 7. Voss, M., Schröder, B. & Fluhrer, R. Mechanism, specificity, and physiology of signal peptide

peptidase (SPP) and SPP-like proteases. Biochim Biophys Acta 1828, 2828–39 (2013).

8. Fluhrer, R., Steiner, H. & Haass, C. Intramembrane proteolysis by signal peptide peptidases: a comparative discussion of GXGD-type aspartyl proteases. J Biol Chem 284, 13975–9 (2009). 9. Brown, M. S., Ye, J., Rawson, R. B. & Goldstein, J. L. Regulated intramembrane proteolysis: a

control mechanism conserved from bacteria to humans. Cell 100, 391–8 (2000).

10. Brown, M. S. & Goldstein, J. L. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell 89, 331–40 (1997).

11. Castelli, M. et al. Different functions of HOPS isoforms in the cell: HOPS shuttling isoform is determined by RIP cleavage system. Cell Cycle 13, 293–302 (2014).

12. Friedmann, E. et al. SPPL2a and SPPL2b promote intramembrane proteolysis of TNFalpha in activated dendritic cells to trigger IL-12 production. Nat Cell Biol 8, 843–8 (2006).

13. Fluhrer, R. et al. A gamma-secretase-like intramembrane cleavage of TNFalpha by the GxGD aspartyl protease SPPL2b. Nat Cell Biol 8, 894–6 (2006).

14. De Strooper, B. et al. A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain. Nature 398, 518–22 (1999).

15. Lichtenthaler, S. F., Haass, C. & Steiner, H. Regulated intramembrane proteolysis--lessons from amyloid precursor protein processing. J Neurochem 117, 779–96 (2011).

16. Tarkowski, E. et al. Cerebral pattern of pro- and anti-inflammatory cytokines in dementias.

Brain Res Bull 61, 255–60 (2003).

(23)

Chapter 6

18. Schneppenheim, J. et al. The intramembrane proteases Signal-peptide-peptidase-like 2a and b (SPPL2a/b) have distinct functions in vivo. Mol Cell Biol (2014).

19. Eikelenboom, P. et al. Neuroinflammation - an early event in both the history and pathogenesis of Alzheimer’s disease. Neurodegener Dis 7, 38–41 (2010).

20. Vom Berg, J. et al. Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease-like pathology and cognitive decline. Nat Med 18, 1812–9 (2012).

21. Chen, C.-H. et al. Increased NF-κB signalling up-regulates BACE1 expression and its therapeutic potential in Alzheimer’s disease. Int J Neuropsychopharmacol 1–14 (2011). doi:10.1017/ S1461145711000149

22. Gorlovoy, P., Larionov, S., Pham, T. T. H. & Neumann, H. Accumulation of tau induced in neurites by microglial proinflammatory mediators. FASEB J 23, 2502–13 (2009).

23. Black, R. A. et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 385, 729–33 (1997).

24. Tan, M.-S. et al. IL12/23 p40 inhibition ameliorates Alzheimer’s disease-associated neuropathology and spatial memory in SAMP8 mice. J Alzheimers Dis 38, 633–46 (2014). 25. Fotinopoulou, A. et al. BRI2 interacts with amyloid precursor protein (APP) and regulates

amyloid beta (Abeta) production. J Biol Chem 280, 30768–72 (2005).

26. Matsuda, S. et al. The familial dementia BRI2 gene binds the Alzheimer gene amyloid-beta precursor protein and inhibits amyloid-beta production. J Biol Chem 280, 28912–6 (2005). 27. Matsuda, S., Giliberto, L., Matsuda, Y., McGowan, E. M. & D’Adamio, L. BRI2 inhibits amyloid

beta-peptide precursor protein processing by interfering with the docking of secretases to the substrate. J Neurosci 28, 8668–76 (2008).

28. Matsuda, S., Matsuda, Y., Snapp, E. L. & D’Adamio, L. Maturation of BRI2 generates a specific inhibitor that reduces APP processing at the plasma membrane and in endocytic vesicles.

Neurobiol Aging 32, 1400–8 (2009).

29. Matsuda, S., Tamayev, R. & D’Adamio, L. Increased AβPP Processing in Familial Danish Dementia Patients. J Alzheimers Dis 27, 385–91 (2011).

30. Tamayev, R., Matsuda, S., Giliberto, L., Arancio, O. & D’Adamio, L. APP heterozygosity averts memory deficit in knockin mice expressing the Danish dementia BRI2 mutant. EMBO J 30, 2501–9 (2011).

31. Tamayev, R. & D’Adamio, L. Memory deficits of British Dementia knock-in mice are prevented by APP haploinsufficiency. J Neurosci 32, 5481–5485 (2012).

32. Kim, J. et al. BRI2 (ITM2b) inhibits Abeta deposition in vivo. J Neurosci 28, 6030–6 (2008). 33. Peng, S., Fitzen, M., Jörnvall, H. & Johansson, J. The extracellular domain of Bri2 (ITM2B) binds

the ABri peptide (1-23) and amyloid beta-peptide (Abeta1-40): Implications for Bri2 effects on processing of amyloid precursor protein and Abeta aggregation. Biochem Biophys Res

(24)

34. Willander, H. et al. BRICHOS Domains Efficiently Delay Fibrillation of Amyloid β-Peptide. J Biol

Chem 287, 31608–17 (2012).

35. Kilger, E. et al. BRI2 regulates {beta}-amyloid degradation by increasing levels of secreted insulin degrading enzyme (IDE). J Biol Chem 286, 37446–37457 (2011).

36. Tsachaki, M. et al. BRI2 interacts with BACE1 and regulates its cellular levels by promoting its degradation and reducing its mRNA levels. Curr Alzheimer Res 10, 532–41 (2013).

37. Vidal, R. et al. A stop-codon mutation in the BRI gene associated with familial British dementia.

Nature 399, 776–81 (1999).

38. Kim, S. H. et al. Familial British dementia: expression and metabolism of BRI. Ann N Y Acad Sci

920, 93–9 (2000).

39. Tamayev, R. et al. Memory Deficits Due to Familial British Dementia BRI2 Mutation Are Caused by Loss of BRI2 Function Rather than Amyloidosis. J Neurosci 30, 14915–14924 (2010). 40. Tamayev, R., Matsuda, S., Fà, M., Arancio, O. & Adamio, L. D. Danish dementia mice suggest that

loss of function and not the amyloid cascade causes synaptic plasticity and memory deficits.

PNAS 107, 20822–27 (2010).

41. Kim, S. H. et al. Furin mediates enhanced production of fibrillogenic ABri peptides in familial British dementia. Nat Neurosci 2, 984–8 (1999).

42. Endres, K. & Fahrenholz, F. Regulation of alpha-secretase ADAM10 expression and activity. Exp

Brain Res 217, 343–52 (2012).

43. Tyler, S. J., Dawbarn, D., Wilcock, G. K. & Allen, S. J. alpha- and beta-secretase: profound changes in Alzheimer’s disease. Biochem Biophys Res Commun 299, 373–376 (2002).

44. Van der Flier, W. M. et al. Optimizing Patient Care and Research: The Amsterdam Dementia Cohort. J Alzheimers Dis (2014). doi:10.3233/JAD-132306

45. Mulder, C. et al. Amyloid-beta(1-42), total tau, and phosphorylated tau as cerebrospinal fluid biomarkers for the diagnosis of Alzheimer disease. Clin Chem 56, 248–53 (2010).

46. Del Campo, M. et al. Recommendations to standardize preanalytical confounding factors in Alzheimer’s and Parkinson’s disease cerebrospinal fluid biomarkers: an update. Biomark Med

6, 419–30 (2012).

47. Voss, M. et al. Foamy virus envelope protein is a substrate for signal peptide peptidase-like 3 (SPPL3). J Biol Chem 287, 43401–9 (2012).

48. Ross, C. a & Poirier, M. a. Protein aggregation and neurodegenerative disease. Nat Med 10

Suppl, S10–7 (2004).

49. Fernández-Nogales, M. et al. Huntington’s disease is a four-repeat tauopathy with tau nuclear rods. Nat Med 20, 4–10 (2014).

50. García-Ayllón, M.-S. et al. CSF Presenilin-1 complexes are increased in Alzheimer’s disease. Acta

(25)

Chapter 6

51. Friedmann, E. et al. Consensus analysis of signal peptide peptidase and homologous human aspartic proteases reveals opposite topology of catalytic domains compared with presenilins.

J Biol Chem 279, 50790–8 (2004).

52. Thal, D. R., Capetillo-Zarate, E., Del Tredici, K. & Braak, H. The development of amyloid beta protein deposits in the aged brain. Sci Aging Knowledge Environ 6, 1–9 (2006).

53. Braak, H., Alafuzoff, I., Arzberger, T., Kretzschmar, H. & Del Tredici, K. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta

Neuropathol 112, 389–404 (2006).

54. Savva, G. et al. Age, Neuropathology, and Dementia — NEJM. N Engl J Med 2302–09 (2009). at <http://www.nejm.org/doi/full/10.1056/NEJMoa0806142>

55. Nelson, P. T. et al. Clinicopathologic correlations in a large Alzheimer disease center autopsy cohort: neuritic plaques and neurofibrillary tangles “do count” when staging disease severity.

J Neuropathol Exp Neurol 66, 1136–46 (2007).

56. Gold, G. et al. Clinical validity of Braak neuropathological staging in the oldest-old. Acta

Neuropathol 99, 579–82; discussion 583–4 (2000).

57. Tarkowski, E., Andreasen, N., Tarkowski, A. & Blennow, K. Intrathecal inflammation precedes development of Alzheimer’s disease. J Neurol Neurosurg Psychiatry 74, 1200–05 (2003). 58. Del Campo, M. & Teunissen, C. E. Role of BRI2 in dementia. J Alzheimers Dis 40, 481–94 (2014). 59. Krawitz, P. et al. Differential localization and identification of a critical aspartate suggest non-redundant proteolytic functions of the presenilin homologues SPPL2b and SPPL3. J Biol Chem

280, 39515–23 (2005).

60. Ciechanover, A. Proteolysis: from the lysosome to ubiquitin and the proteasome. Nat Rev Mol

Cell Biol 6, 79–87 (2005).

61. Chambraud, B. et al. A role for FKBP52 in Tau protein function. Proc Natl Acad Sci U S A 107, 2658–63 (2010).

62. Luong, a, Hannah, V. C., Brown, M. S. & Goldstein, J. L. Molecular characterization of human acetyl-CoA synthetase, an enzyme regulated by sterol regulatory element-binding proteins. J

Biol Chem 275, 26458–66 (2000).

63. Voss, K., Combs, B., Patterson, K., Binder, L. I. & Chris, T. Hsp70 alters tau function and aggregation in an isoform specific manner. Biochemistry 51, 888–898 (2013).

64. Lee, M. J., Lee, J. H. & Rubinsztein, D. C. Tau degradation: the ubiquitin-proteasome system versus the autophagy-lysosome system. Prog Neurobiol 105, 49–59 (2013).

65. Street, J. M. et al. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med 10, 5 (2012).

(26)

67. Schrul, B., Kapp, K., Sinning, I. & Dobberstein, B. Signal peptide peptidase (SPP) assembles with substrates and misfolded membrane proteins into distinct oligomeric complexes. Biochem J

427, 523–34 (2010).

68. Friedmann, E. et al. Protein Structure and Folding : Consensus Analysis of Signal Peptide Peptidase and Homologous Human Aspartic Proteases Reveals Opposite Topology of Catalytic Domains Compared with Presenilins Consensus Analysis of Signal Peptide Peptidase and Homologous H. J Biol Chem 50790–98 (2004). doi:10.1074/jbc.M407898200

69. Motter, R. et al. Reduction of beta-amyloid peptide42 in the cerebrospinal fluid of patients with Alzheimer’s disease. Ann Neurol 38, 643–8 (1995).

70. Jack, C. R. et al. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet, The 9, 1–20 (2010).

71. Navarro, A., Del Valle, E., Astudillo, A., González del Rey, C. & Tolivia, J. Immunohistochemical study of distribution of apolipoproteins E and D in human cerebral beta amyloid deposits. Exp

Neurol 184, 697–704 (2003).

(27)

Referenties

GERELATEERDE DOCUMENTEN

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded

The molecular role of Serf2 in development and misfolded protein aggregation Stroo, Esther.. IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright