the amyloid precursor protein forms plasmalemmal clusters via its pathogenic amyloid-β domain

7
The Amyloid Precursor Protein Forms Plasmalemmal Clusters via Its Pathogenic Amyloid-b Domain Arne Schreiber, Sebastian Fischer, and Thorsten Lang * Department of Membrane Biochemistry, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany ABSTRACT The amyloid precursor protein (APP) is a large, ubiquitous integral membrane protein with a small amyloid- b (Ab) domain. In the human brain, endosomal processing of APP produces neurotoxic Ab-peptides, which are involved in Alzheimer’s disease. Here, we show that the Ab sequence exerts a physiological function when still present in the unpro- cessed APP molecule. From the extracellular site, Ab concentrates APP molecules into plasmalemmal membrane protein clusters. Moreover, Ab stabilization of clusters is a prerequisite for their targeting to endocytic clathrin structures. Therefore, we conclude that the Ab domain directly mediates a central step in APP trafficking, driving its own conversion into neurotoxic peptides. INTRODUCTION The human amyloid precursor protein (APP) gene encodes for a ubiquitously expressed type I integral membrane protein with a large extracellular domain and a short cyto- plasmic region (1). Three major isoforms, ranging from 695 to 770 amino acids (aa) in length, have been identified. APP first attracted attention when one of its degradation products, the ~40-aa large b-amyloid (Ab) peptide, was found in senile brain plaques isolated from Alzheimer’s patients (2). Though the primary function of APP still remains elusive, most research focuses on the function and generation of the Ab cleavage product. To date, it is clear how Ab peptides are generated and also that alternative APP degradation pathways exist. For Ab production, the enzymes b- and g-secretase are required to cleave the Ab sequence at its N- and C-terminal ends, respectively (3). As b-secretase functions in early endo- somes (4–6), endocytic uptake of APP molecules is a prereq- uisite for amyloidogenic processing. Alternatively, the Ab region is cleaved at its center by plasmalemmal a-secretase, yielding harmless fragments (nonamyloidogenic pathway). After processing and secretion of Ab peptides, Ab oligo- mers or fibrils play a central role in the development of Alzheimer’s disease (7). Moreover, several nonpathogenic activities of Ab peptides have been discussed (for example, see Baruch-Suchodolsky et al. (8), Igbavboa et al. (9), and Zou et al. (10)). However, so far it is unknown whether the Ab domain has a function when still present in the unpro- cessed molecule (for exception, see Tienari et al. (11)). Interfering with trafficking or processing of APP might reduce the progression of the disease (12,13), for instance, by taking advantage of a- and b-secretase being active at the plasma membrane and in early endosomes, respectively. A block of APP endocytosis would favor a-processing and at the same time avoid b-cleavage, resulting in overall non- amyloidogenic processing (14). Internalization of APP occurs via a clathrin-mediated pathway (15). Moreover, it is known that endocytosis of APP is regulated by a flotillin-dependent APP clustering step (16). However, flotillin rather acts as a modulator of a more intrinsic clustering mechanism, since clusters become smaller but still remain after flotillin downregulation (16). Here, we examine the mechanism of APP clustering in the plasma membrane, as understanding APP clustering is important for understanding APP trafficking, which in turn is a prerequisite for the development of therapeutic strate- gies against Alzheimer’s disease. MATERIALS AND METHODS Cloning of APP constructs The expression vector used for all constructs was pcDNA6.2/C-emGFP/ DEST (V355-20, Invitrogen, Carlsbad, CA). N-terminally myc-tagged human APP695 (NCBI reference sequence NM_201414 with VPEQKLISEEDL inserted between positions 19 and 20 (11,17)) was inserted via PCR/Gateway cloning (Invitrogen) into the expression vector, resulting in C-terminally GFP-tagged myc-APP with a KGGRADPAFLYKVVDAVN linker between APP and GFP. Following the same procedure, a C-terminally deleted construct was obtained (APP-DC; lacking aa 649–695 of the original APP sequence). For N-terminal deletion, a cDNA sequence was obtained from MWG Eurofins (Ebersberg, Germany) and inserted via PCR/Gateway cloning into the expres- sion vector (APP-DN; lacking aa 18–601). The myc-tagged full-length APP was used in Figs. 1 and 2, Fig. S1, Fig. S2, and Fig. S4 C. For Figs. 3–5, Fig. S3, Fig. S5, and Fig. S6, we used APP without myc-tag that was cloned by using the Acc65I and EcoRI restriction sites within the APP sequence. To this end, a double-digested PCR product was inserted into the double-digested pcDNA6.2/C-emGFP/DEST expression vector from the myc-tagged APP, resulting in a construct without the VPEQKLISEEDL insert. From this plasmid, using the restriction sites Acc65I and EcoRI or Acc65I and SgsI, deletion constructs were produced (the deletions D22–283, D22–435, D22–537, D22–596, and D22–601). In addition, fusion PCR products carrying the mutations N467S and/or T291G, T292G and T576G were amplified and inserted into the expression vector. Using the NCBI reference sequence NM_201414 as reference, the APP coding sequence of all constructs was verified by sequencing. Comparative Submitted January 11, 2012, and accepted for publication February 21, 2012. *Correspondence: [email protected] Editor: Petra Schwille. Ó 2012 by the Biophysical Society 0006-3495/12/03/1411/7 $2.00 doi: 10.1016/j.bpj.2012.02.031 Biophysical Journal Volume 102 March 2012 1411–1417 1411

Upload: thorsten

Post on 25-Nov-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Biophysical Journal Volume 102 March 2012 1411–1417 1411

The Amyloid Precursor Protein Forms Plasmalemmal Clusters via ItsPathogenic Amyloid-b Domain

Arne Schreiber,† Sebastian Fischer,† and Thorsten Lang†*†Department of Membrane Biochemistry, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany

ABSTRACT The amyloid precursor protein (APP) is a large, ubiquitous integral membrane protein with a small amyloid-b (Ab) domain. In the human brain, endosomal processing of APP produces neurotoxic Ab-peptides, which are involved inAlzheimer’s disease. Here, we show that the Ab sequence exerts a physiological function when still present in the unpro-cessed APP molecule. From the extracellular site, Ab concentrates APP molecules into plasmalemmal membrane proteinclusters. Moreover, Ab stabilization of clusters is a prerequisite for their targeting to endocytic clathrin structures. Therefore,we conclude that the Ab domain directly mediates a central step in APP trafficking, driving its own conversion into neurotoxicpeptides.

INTRODUCTION

The human amyloid precursor protein (APP) gene encodesfor a ubiquitously expressed type I integral membraneprotein with a large extracellular domain and a short cyto-plasmic region (1). Three major isoforms, ranging from695 to 770 amino acids (aa) in length, have been identified.APP first attracted attention when one of its degradationproducts, the ~40-aa large b-amyloid (Ab) peptide, wasfound in senile brain plaques isolated from Alzheimer’spatients (2). Though the primary function of APP stillremains elusive, most research focuses on the function andgeneration of the Ab cleavage product.

To date, it is clear how Ab peptides are generated and alsothat alternative APP degradation pathways exist. For Abproduction, the enzymes b- and g-secretase are requiredto cleave the Ab sequence at its N- and C-terminal ends,respectively (3). As b-secretase functions in early endo-somes (4–6), endocytic uptake of APPmolecules is a prereq-uisite for amyloidogenic processing. Alternatively, the Abregion is cleaved at its center by plasmalemmal a-secretase,yielding harmless fragments (nonamyloidogenic pathway).

After processing and secretion of Ab peptides, Ab oligo-mers or fibrils play a central role in the development ofAlzheimer’s disease (7). Moreover, several nonpathogenicactivities of Ab peptides have been discussed (for example,see Baruch-Suchodolsky et al. (8), Igbavboa et al. (9), andZou et al. (10)). However, so far it is unknown whether theAb domain has a function when still present in the unpro-cessed molecule (for exception, see Tienari et al. (11)).

Interfering with trafficking or processing of APP mightreduce the progression of the disease (12,13), for instance,by taking advantage of a- and b-secretase being active atthe plasma membrane and in early endosomes, respectively.A block of APP endocytosis would favor a-processing and

Submitted January 11, 2012, and accepted for publication February 21,

2012.

*Correspondence: [email protected]

Editor: Petra Schwille.

� 2012 by the Biophysical Society

0006-3495/12/03/1411/7 $2.00

at the same time avoid b-cleavage, resulting in overall non-amyloidogenic processing (14).

Internalization of APP occurs via a clathrin-mediatedpathway (15). Moreover, it is known that endocytosis ofAPP is regulated by a flotillin-dependent APP clusteringstep (16). However, flotillin rather acts as a modulator of amore intrinsic clustering mechanism, since clusters becomesmaller but still remain after flotillin downregulation (16).

Here, we examine the mechanism of APP clustering inthe plasma membrane, as understanding APP clustering isimportant for understanding APP trafficking, which in turnis a prerequisite for the development of therapeutic strate-gies against Alzheimer’s disease.

MATERIALS AND METHODS

Cloning of APP constructs

The expression vector used for all constructs was pcDNA6.2/C-emGFP/

DEST(V355-20, Invitrogen,Carlsbad,CA).N-terminallymyc-taggedhuman

APP695 (NCBI reference sequence NM_201414 with VPEQKLISEEDL

inserted between positions 19 and 20 (11,17)) was inserted via PCR/Gateway

cloning (Invitrogen) into the expression vector, resulting in C-terminally

GFP-tagged myc-APP with a KGGRADPAFLYKVVDAVN linker between

APPandGFP.Following the sameprocedure, aC-terminally deleted construct

was obtained (APP-DC; lacking aa 649–695 of the original APP sequence).

ForN-terminal deletion, a cDNAsequencewas obtained fromMWGEurofins

(Ebersberg, Germany) and inserted via PCR/Gateway cloning into the expres-

sion vector (APP-DN; lacking aa 18–601). The myc-tagged full-length APP

was used in Figs. 1 and 2, Fig. S1, Fig. S2, and Fig. S4 C. For Figs. 3–5,

Fig. S3, Fig. S5, and Fig. S6, we used APP without myc-tag that was cloned

by using the Acc65I and EcoRI restriction sites within the APP sequence. To

this end, a double-digested PCRproductwas inserted into the double-digested

pcDNA6.2/C-emGFP/DEST expression vector from the myc-tagged APP,

resulting in a construct without the VPEQKLISEEDL insert. From this

plasmid, using the restriction sites Acc65I and EcoRI or Acc65I and SgsI,

deletion constructs were produced (the deletions D22–283, D22–435,

D22–537,D22–596, andD22–601). In addition, fusionPCRproducts carrying

the mutations N467S and/or T291G, T292G and T576G were amplified and

inserted into the expression vector.

Using the NCBI reference sequence NM_201414 as reference, the APP

coding sequence of all constructs was verified by sequencing. Comparative

doi: 10.1016/j.bpj.2012.02.031

FIGURE 1 APP mobility is restricted via the

N-terminal extracellular domain. (A) Confocal

micrographs were recorded from PC12 cells ex-

pressing GFP-tagged APP (upper) and variants

with an N-terminal deletion (APP-DN; lower) or

a C-terminal deletion (APP-DC; not shown). Fluo-

rescence bleaching was performed in a squared

ROI (dotted square) at the basal plasma membrane

(compare first and second images). Recovering

fluorescence was related to the prebleach value.

For one experiment, normalized recovery traces

from several cells were averaged for calculation

of the half-time of recovery. (B and C) Averaged

experiments. Values are given as the mean 5 SE

(n ¼ 3–5).

1412 Schreiber et al.

fluorescence recovery after photobleaching (FRAP) analysis showed no

difference between the two full-length APP constructs (with and without

myc-tag; data not shown).

Cell culture

PC12 cells were maintained, propagated, and plated onto poly-L-lysine

coated coverslips essentially as previously described (18). HepG2 cells

were propagated in EMEM (BE12-992F, Lonza, Basel, Switzerland) supple-

mentedwith 10%fetal calf serumand1%Pen/Strep at 37�Cand5%CO2. For

transfection, we used a Microporator MP100 (Digital Bio, Seoul, South

Korea) or a Neon-Transfection System (Invitrogen) applying 100-ml gold

tips, adding 10–25 mg plasmid/transfection tip and adjusting settings to

1410 V and 30 ms for PC12 cells or 1200 V and 50 ms for HepG2 cells.

For Fig. S4, cells were treated before the FRAP experiment with deglycosy-

lating enzymes a-neuraminidase (0.5 U/ml) and peptide-N-glycosidase

(0.1 U/ml) (N2876 and G5166, respectively, Sigma, St. Louis, MO) and

b-galactosidase (0.16 U/ml; P07305, New England Biolabs, Ipswich, MA)

in serum-free medium for 1 h at 37�C in the cell incubator. Then cells

were mounted into a microscopy chamber in Ringer solution (130 mM

FIGURE 2 APP disperses after deletion of the extracellular domain.

TIRF microscopy images from PC12 cells expressing the same constructs

as in Fig. 1, illustrating that APP disperses upon deletion of the extracellular

domain (APP-DN), whereas it remains clustered without the intracellular

C-terminal domain (APP-DC). Images are shown at arbitrary scaling.

Biophysical Journal 102(6) 1411–1417

NaCl, 4 mM KCl, 1 mM CaCl2, 1 mM MgCl2, 48 mM glucose, and

10 mM HEPES, pH 7.4). Cells were used ~8 h (Figs. 3–5, Fig. S3, Fig. S4,

Fig. S5, and Fig. S6), 24 h (Figs. 1 and 2 and Fig. S2), and 8–24 h

(Fig. S1) after transfection.

FRAP analysis

FRAP measurements in Ringer solution at room temperature (RT) and

analyses were performed using an Olympus Fluo View 100 laser scanning

microscope essentially as described previously (18). In brief, the pixel size

was adjusted to 207 nm and the image size to 100� 100 pixel, and bleaching

was performed in a 15� 15 pixel region of interest (ROI) using the 488-nm

and 405-nm laser lines simultaneously. Depending on the experiment,

bleaching duration varied from 1–2 s. In total, a sequence of 70 images

was recorded, including three prebleach and 67 postbleach images. For anal-

ysis, apart from the bleaching ROI, values were obtained from a reference

ROI in an unbleached area and a background value was determined.

Recovery traces were background-subtracted and normalized to the average

of the prebleach values. Vertical drift during the measurement was indicated

by a loss of fluorescence in the reference ROI. When the value of the refer-

ence ROI decreased by >15%, the measurement was excluded.

The half-time of recovery was obtained by fitting a hyperbola (y(t)¼ off-

set þ maxrec � t/(t þ t1/2)) using Origin8Pro (OriginLab, Northampton,

MA). For one experiment and condition, 3–11 cells were analyzed and

half-times and normalized recovery traces were averaged.

The diffusion coefficient of a freely diffusing single-span membrane

protein has been reported to be 0.23 mm2/s (19), which corresponds to

the value 0.13 of the normalized half-time (Fig. 3).

TIRF microscopy

Total internal reflection fluorescence (TIRF) microscopy was performed

essentially as already described (18). Cells were imaged at RT in Ringer

solution (n ¼ 3–4 independent experiments).

FIGURE 3 Entire Ab domain is essential for mobility restriction of APP.

(A) FRAP recovery traces (as in Fig. 1 B) from APP constructs carrying the

indicated deletions within the ectodomain. (B) Half-times were normalized

to full-length APP and plotted against the number of deleted aa. The shad-

owed area indicates the half-time zone for a freely diffusing single-span

membrane protein (see also Materials and Methods). The erratic increase

in mobility from construct D22–596 to D22–601 indicates a change from

restricted to free diffusion. Please note that the two constructs differ by

only five aa from the N-terminus of the amyloid b region. (Upper) Domain

structure of APP (not to scale; referring to Reinhard et al. (29)). Dashed

lines roughly indicate the position of the last deleted amino acid of the

respective construct. GFLD, N-terminal growth-factor-like domain;

CuBD, copper-binding domain; AcD, acidic domain; RERMS, five-aa

peptide sequence; CAPPD, central APP domain; L, linker; Ab, amyloid-b

peptide sequence; AICD, APP intracellular domain. Values are given as the

mean 5 SE (n ¼ 3–12).

FIGURE 4 Truncation of Ab is accompanied by cluster dispersion. Fixed

membrane sheets generated from cells expressing the constructs as indi-

cated. From individual membrane sheets the SD of the mean intensity

was determined in a ROI and related to the mean background-corrected

fluorescence, yielding the relative SD, which is a quantitative measure

for the degree of clustering (18). For each construct, data from three to

four independent experiments were pooled. From individual membrane

sheets, the relative SD was plotted against the mean intensity and a function

f(x) ¼ a�xb was fitted (APP, a ¼ 0.9961 and b ¼ �0.295; APPD22–596,

a ¼ 1.169 and b ¼ �0.321; APPD22–601, a ¼ 0.7785 and b ¼ �0.295).

Please note that maximal and minimal values obtained at low and high

expression levels are a consequence of mathematical processing and do

not necessarily indicate that clusters disappear at high expression levels

(the same effect is expected if clusters just become more numerous).

Full-length APP (blue line) and APPD22–596 (red line) have the same

degree of clustering, whereas the predominantly uniform distribution of

APPD22–601 (green line) is indicated by a shift of the fitted line toward

lower values. For each condition, 62–105 individual membrane sheets

were analyzed. Images are shown at arbitrary scaling.

APP Clustering via the Amyloid-b Domain 1413

Wheat germ agglutinin staining

For Fig. S4, cells were pretreated with deglycosylating enzymes as above.

Then membrane sheets were generated (18) and fixed with 4% paraformal-

dehyde (PFA) in phosphate-buffered saline (PBS; 137 mM NaCl, 2.7 mM

KCl, and 8.1 mM Na2HPO4, pH7.4) for 1 h. PFA was quenched with

50 mM NH4Cl in PBS and membrane sheets were incubated with 5 mg/ml

Alexa594-labeled wheat germ agglutinin (WGA) (W11262, Invitrogen) in

PBS. Membrane sheets were washed with PBS and imaged in PBS contain-

ing 1-(4-tri-methyl-ammonium-phenyl)-6-phenyl-1,3,5-hexatriene p-tolue-

nesulfonate (TMA-DPH) (T204, Molecular Probes, Eugene, OR). Imaging

and image analysis (using the program ImageJ) were performed as

described (18). For each independent experiment and condition, 24–39

membrane sheets were analyzed and values were averaged.

Immunostaining of membrane sheets

Immunostaining was performed according to a protocol essentially as

previously described (18). For clathrin staining, a goat antibody directed

against the clathrin heavy chain (sc-6579, Santa Cruz Biotechnology, Santa

Cruz, CA) and, as secondary antibody, a donkey-anti-goat antibody coupled

to Alexa594 (A11058, Invitrogen) were used. Membrane sheets were

imaged in the presence of TMA-DPH to illustrate the integrity of the

membrane. Membrane sheets with the expressed constructs were identified

in the green channel and then imaged in the blue, green, and red channels to

show the lipid staining (TMA-DPH fluorescence), GFP-fluorescence, and

clathrin staining, respectively.

To correct for lateral shifts occurring during channel switching, and to

ensure that the focus was properly adjusted, we added 100 nm TetraSpeck

beads (T7279, Invitrogen/Molecular Probes) to the coverslip that were

detectable in all channels. The correlation coefficient was determined

using ImageJ, applying the ImageJ plugin Colocalization Indices (20).

For each independent experiment and condition, 4–14 membrane sheets

were analyzed and their values were averaged. For full-length APP, we

also analyzed the overlap of APP and clathrin on the level of individual

Biophysical Journal 102(6) 1411–1417

FIGURE 5 Targeting of APP into clathrin structures requires the entire

Ab- and the intracellular domain. Membrane sheets from PC12 cells ex-

pressing GFP-tagged APP, APPD22–283 (not shown), APPD22–596,

APPD22–601, or APP-DC were fixed and immunostained for clathrin.

Shown from left to right are TMA-DPH staining visualizing the integrity

of the plasma membrane; the GFP channel showing the distribution of

the respective APP construct; clathrin staining indicating endocytic clathrin

structures and an overlay of GFP/clathrin signals. From central areas, the

correlation between the GFP and the clathrin staining patterns was quanti-

fied by calculation of the Pearson correlation coefficient. The coefficient

yields 1 in the case of two identical pictures (realistic values are much lower

due to technical reasons), 0 when signals are unrelated, and �1 in the case

of a picture and its negative. On the level of individual spots, the value of

0.32 obtained for full-length APP corresponds to 54% of APP clusters

that are positive for clathrin (Fig. S6). Values are given as the mean 5 SE

(n ¼ 3–4). Images are shown at arbitrary scaling.

1414 Schreiber et al.

spots (Fig. S6) and corrected for accidental colocalization as previously

described (21).

Detection of constructs at the cell surface

For detection of GFP-labeled APP, APPD22–596, and APPD22–601 at the

cell surface, we followed a protocol essentially as previously described

Biophysical Journal 102(6) 1411–1417

(16). In brief, PC12 cells expressing the respective constructs were cooled

down in the cold room on ice, and all the steps described here were per-

formed with ice-cold solutions. Cells were washed with Hank’s BSS

(HBSS; cat# H15-008, PAA Laboratories, Colbe, Germany) and incubated

for 30 min with a mouse monoclonal primary antibody (1:100 in HBSS;

SIG-39300, Covance, Princeton, NJ) reactive for aa 1–16 of the Ab peptide.

Then cells were washed and incubated for 40 min with secondary antibody

donkey-anti-mouse coupled to Alexa594 (1:200 in HBSS; A21203, Invitro-

gen). Cells were washed and fixed overnight in 4% PFA in PBS. Quenching

of PFAwith NH4Cl and embedding in Mowiol (6 g glycerol, 2.4 g Mowiol

(4-88, Hoechst, Frankfurt, Germany), 6 ml ddH2O, and 12 ml of 200 mM

Tris, pH 7.2) saturated with Dabco (0718.1, Carl Roth, Karlsruhe,

Germany) was performed at RT. For imaging, we used the Olympus Fluo

View 100 laser scanning microscope, applying the laser line 488 for GFP

and the laser line 543 for Alexa594. In addition, cells were imaged in

the differential interference contrast (DIC) mode. For analysis, we used

ImageJ. In brief, using the DIC image as reference, a linescan was placed

at the rim of the cell. In the green (GFP) and red (immunostaining) channels,

the average intensities per pixel were measured and background-corrected.

Pooling data from several independent experiments, immunostaining

intensity was plotted against GFP fluorescence for each cell.

RESULTS AND DISCUSSION

Clustered membrane proteins are restricted in diffusion (see,e.g., Zilly et al. (18) and He and Marguet (22)). Therefore, toprobe the clustered state of APP molecules, we analyzed itsdiffusional mobility by FRAP. For fluorescent labeling, GFPwas fused to the C terminus of the 695-aa large neuronalAPP isoform and the construct was expressed in neuronaland liver model cell lines. For FRAP analysis, we focusedon the basal plasma membrane, bleached fluorescence ina squared ROI, and monitored the recovery of fluorescenceover time. The recovery rate was much slower than expectedfor free diffusion, although it was somewhat dependent onthe time after expression, becoming accelerated between8 and 24 h after transfection (Fig. S1). To test which proteindomains restrict mobility in neuronal cells, we analyzeddiffusion of a deletion construct lacking the complete extra-cellular domain (APP-DN) and found a severalfold highermobility when compared to full-length APP (Fig. 1). Wethen asked whether this increased mobility is indeed dueto a change from clustered to uniform molecule organiza-tion. To this end, we applied TIRF microscopy, which selec-tively illuminates the plasma membrane region, avoidingthe imaging of deeper-lying intracellular compartments.As shown in Fig. 2, the majority of APP fluorescencewas concentrated into punctate structures, whereas uponremoval of the ectodomain (APP-DN) the distributionbecame uniform with additional spotty signals occasionallyseen. This is in line with our assumption that fast and slowdiffusion in FRAP experiments correspond to low and highdegrees of protein clustering, respectively. Next, we deletedthe intracellular domain (construct APP-DC, with the fullextracelluar domain intact). The mobility was slightlyincreased (Fig. 1, B and C), and the distribution of APP-DCstill had a clustered appearance in TIRF micrographs,although it was less clear than for APP (Fig. 2). Analysis

APP Clustering via the Amyloid-b Domain 1415

of the deletion constructs in liver cells (Fig. S2) revealedidentical results, implying that neuronal factors are notinvolved in the basic APP clustering mechanism.

So far, the data suggest that the extracellular domain ofAPP is essential for concentrating the protein into clustersin the membrane. In the absence of the intracellular domain,clusters still form but become more dynamic, in line withthe presence of modulators stabilizing APP clusters fromthe intracellular side.

The extracellular domain could mediate APP clusteringby several mechanisms. For instance, APP contains glyco-sylation sites in the ectodomain (23,24) that could interactwith or be part of the extracellular glycocalyx. Other possi-bilities are that the bulkiness of the ectodomain couldrestrict mobility, or a special protein domain could beresponsible for clustering. To test the first possibility, weperformed FRAP experiments on cells with APP harboringpoint mutations at the glycosylation sites (Fig. S3) or cellswith a degraded glycocalyx (Fig. S4). However, APPdiffused normally, ruling out a role of the glycocalyx. Wethen performed FRAP analysis with progressively shorterconstructs (Fig. 3). Over awide sequence range, the deletionscaused only small mobility enhancements approximatelylinear to the number of removed aa. This observationmight be due to friction forces, which can be expected tobe reduced with the smaller geometric sizes of the extracel-lular domains. In any case, because deletion of almostthe entire ectodomain (construct APPD22–537) raised themobility only modestly (Fig. 3), we can conclude that theglobular size of the ectodomain does not produce a clusteringpattern due to restriction by extracellular diffusion barriers(for a suggested mechanism based on intracellular diffu-sion barriers, see Kusumi et al. (25)). Notably, the mobilitychanged upon further deletion, with a remarkable stepwiseincrease caused by removing the first five aa of the Abregion. These findings strongly suggest that APP clusteringismediated primarily through a specific interaction involvingthe N-terminal region of the Ab domain, although somestabilizing effect of the more upstream-lying aa is notable.To verify that the protein disperses upon Ab-shortening, wecompared the lateral distribution of APP to the distributionsof the two shortest variants, APPD22–596 andAPPD22–601,applying fixed plasma membrane sheets (Fig. 4). Plasmamembrane sheets were generated by brief ultrasound treat-ment of glass-adhered cells, leaving behind the basal plasmamembrane. This preparation allows for high signal/noiseratio analysis of fluorescence signals. Moreover, unlikeTIRF microscopy, it can be used to exclude the possibilitythat uniform fluorescence results from cytosolic GFP orthat punctate signals arise from undocked vesicles close tothemembrane (see also Fig. S5 for a surface staining showingthe presence of the constructs within the plasma membrane).As shown in Fig. 4, both APP and APPD22–596 show astrongly clustered pattern, whereas APPD22–601 is predom-inantly uniformly distributed. This corroborates the conclu-

sions drawn from the FRAP experiments, that the first aaof the Ab-region dramatically affect the lateral organizationof APP by immobilizing APP into clusters.

We then asked if clustering is an event associated withendocytosis and compared the APP clustering pattern tothe distribution of clathrin. A high degree of overlap wasmeasured only for constructs possessing both the Ab andthe intracellular domain, though the still clustered, butalready more dynamic, APPD22–596 less efficiently corre-lated with clathrin. Compared to full-length APP, theconstruct with the partially deleted Ab domain was almostfivefold less targeted to endocytic structures. This indicatesthat a certain degree of cluster stability is required for effi-cient entry into the endocytic pathway. We also tested theconstruct lacking the intracellular domain (APP-DC). As ex-pected, APP-DC did not incorporate into clathrin structures,instead even showing some trend toward being excludedfrom the clathrin staining pattern (Fig. 5).

In summary, the data show that a few aa within the Abregion are sufficient for the formation of APP clusters. Incontrast, the intracellular domain of APP is incapable offorming clusters on its own, though it gives additional stabi-lization to Ab-formed clusters from the intracellular site.

As shown by our clathrin staining experiments, the intra-cellular portion of APP directs the molecule into endocyticstructures. At present, it is unclear whether clathrin is re-cruited to the site of clustering or whether clustered APPbecomes trapped into clathrin assemblies, but we can safelyconclude that the process is only effective upon APP immo-bilization by Ab. Probably, clathrin structures interfere withthe dynamics of APP clusters, explaining the slight increasein APP mobility upon removal of the intracellular domain.In any case, the data show that tight clustering is a prerequi-site for directing APP efficiently to the endocytic pathway.Therefore, we conclude that APP clustering and immobili-zation is a key step in APP trafficking from the plasmamembrane via clathrin-coated vesicles to early endosomes(Fig. 6).

Inhibition of b-secretase function has already been sug-gested (13,26) as a means to prevent Alzheimer’s disease,but it would require a pharmaceutical with intracellularactivity and would carry the risk of possible side effectsdue to inhibition of an enzyme that also has physiologicalrelevance (27). In contrast, prevention of APP clusteringinto endocytic structures requires the pharmaceutical toact only from the extracellular site (Fig. 6). Moreover, inanalogy to a mechanism suggested to produce shorter Abpeptides when interfering with GxxxG-motif-mediatedAPP dimerization (28), dispersed APP may allow g-secre-tase to bind the APP molecule farther upstream, leading tothe production of shorter and less harmful Ab peptides.

In a nutshell, our data show that for APP clustering, theAb-domain is essential. Moreover, clustering is a prerequi-site for targeting APP to endocytic structures. These find-ings indicate what we consider a novel central step in APP

Biophysical Journal 102(6) 1411–1417

FIGURE 6 APP cluster formation within the plasma membrane. Unpro-

cessed APP molecules reach the cell surface through the constitutive secre-

tory pathway, where they cluster via the N-terminal aa of Ab. APP dimers

may form via alternative mechanisms involving regions of the extracellular

domain (30–32) or consecutive GxxxG motifs more downstream of the Ab

N-terminus (28). With increasing cluster size, APP molecules become

immobile and locally provide more binding sites for the endocytic

machinery, favoring full clathrin assembly or sorting to clathrin structures,

internalization, and amyloidogenic processing. Interference with the clus-

tering reaction from the extracellular site would increase the time APP

resides in the plasma membrane and thereby the chance to be cleaved by

a-secretase, resulting in nonamyloidogenic processing.

1416 Schreiber et al.

trafficking, which is intriguing for two reasons: first, theAb domain initiates its own intracellular production, andsecond, the finding is medically relevant, as it suggests anew route for battling Alzheimer’s disease.

SUPPORTING MATERIAL

Six figures are available at http://www.biophysj.org/biophysj/supplemental/

S0006-3495(12)00262-7.

The authors acknowledge a plasmid gift encoding for myc-tagged human

APP695 from Dr. Stefan Kins (Zentrum fur Molekulare Biologie der Uni-

versitat Heidelberg, Heidelberg, Germany) and thank Dr. Michael Famulok

(University of Bonn, Bonn, Germany) for providing HepG2 cells and

Dr. Thomas Quast for support with the confocal microscope. We also thank

Dr. Ines Raschke, Dr. Jan-Gero Schloetel, and David Walrafen for helpful

comments on the manuscript.

This project was supported by a grant from the Deutsche Forschungsge-

meinschaft (SFB645).

REFERENCES

1. Li, Q. X., S. J. Fuller, ., C. L. Masters. 1999. The amyloid precursorprotein of Alzheimer disease in human brain and blood. J. Leukoc. Biol.66:567–574.

2. Walsh, D. M., and D. J. Selkoe. 2007. A b oligomers—a decade ofdiscovery. J. Neurochem. 101:1172–1184.

Biophysical Journal 102(6) 1411–1417

3. O’Brien, R. J., and P. C. Wong. 2011. Amyloid precursor protein pro-cessing and Alzheimer’s disease. Annu. Rev. Neurosci. 34:185–204.

4. He, X., K. Cooley, ., J. Tang. 2007. Apolipoprotein receptor 2 andX11 a/b mediate apolipoprotein E-induced endocytosis of amyloid-b precursor protein and b-secretase, leading to amyloid-b production.J. Neurosci. 27:4052–4060.

5. Kinoshita, A., H. Fukumoto,., B. T. Hyman. 2003. Demonstration byFRET of BACE interaction with the amyloid precursor protein at thecell surface and in early endosomes. J. Cell Sci. 116:3339–3346.

6. Rajendran, L., M. Honsho, ., K. Simons. 2006. Alzheimer’s diseaseb-amyloid peptides are released in association with exosomes. Proc.Natl. Acad. Sci. USA. 103:11172–11177.

7. Sakono, M., and T. Zako. 2010. Amyloid oligomers: formation andtoxicity of Ab oligomers. FEBS J. 277:1348–1358.

8. Baruch-Suchodolsky, R., and B. Fischer. 2009. Ab40, either soluble oraggregated, is a remarkably potent antioxidant in cell-free oxidativesystems. Biochemistry. 48:4354–4370.

9. Igbavboa, U., G. Y. Sun, ., W. G. Wood. 2009. Amyloid b-proteinstimulates trafficking of cholesterol and caveolin-1 from the plasmamembrane to the Golgi complex in mouse primary astrocytes. Neuro-science. 162:328–338.

10. Zou, K., J. S. Gong, ., M. Michikawa. 2002. A novel function ofmonomeric amyloid b-protein serving as an antioxidant moleculeagainst metal-induced oxidative damage. J. Neurosci. 22:4833–4841.

11. Tienari, P. J., B. De Strooper, ., C. G. Dotti. 1996. The b-amyloiddomain is essential for axonal sorting of amyloid precursor protein.EMBO J. 15:5218–5229.

12. Cirrito, J. R., J. E. Kang, ., D. M. Holtzman. 2008. Endocytosis isrequired for synaptic activity-dependent release of amyloid-b in vivo.Neuron. 58:42–51.

13. Vassar, R., D. M. Kovacs, ., P. C. Wong. 2009. The b-secretaseenzyme BACE in health and Alzheimer’s disease: regulation, cellbiology, function, and therapeutic potential. J. Neurosci. 29:12787–12794.

14. Carey, R. M., B. A. Balcz,., B. E. Slack. 2005. Inhibition of dynamin-dependent endocytosis increases shedding of the amyloid precursorprotein ectodomain and reduces generation of amyloid b protein.BMC Cell Biol. 6:30.

15. Nordstedt, C., G. L. Caporaso,., P. Greengard. 1993. Identification ofthe Alzheimer b/A4 amyloid precursor protein in clathrin-coated vesi-cles purified from PC12 cells. J. Biol. Chem. 268:608–612.

16. Schneider, A., L. Rajendran, ., M. Simons. 2008. Flotillin-dependentclustering of the amyloid precursor protein regulates its endocytosisand amyloidogenic processing in neurons. J. Neurosci. 28:2874–2882.

17. Soba, P., S. Eggert,., K. Beyreuther. 2005. Homo- and heterodimeri-zation of APP family members promotes intercellular adhesion.EMBO J. 24:3624–3634.

18. Zilly, F. E., N. D. Halemani,., T. Lang. 2011. Ca2þ induces clusteringof membrane proteins in the plasma membrane via electrostatic inter-actions. EMBO J. 30:1209–1220.

19. Kenworthy, A. K., B. J. Nichols, ., J. Lippincott-Schwartz. 2004.Dynamics of putative raft-associated proteins at the cell surface.J. Cell Biol. 165:735–746.

20. Nakamura, K., A. Watakabe, ., T. Kaneko. 2007. Transientlyincreased colocalization of vesicular glutamate transporters 1 and 2at single axon terminals during postnatal development of mouseneocortex: a quantitative analysis with correlation coefficient. Eur. J.Neurosci. 26:3054–3067.

21. Lang, T., M. Margittai, ., R. Jahn. 2002. SNAREs in native plasmamembranes are active and readily form core complexes with endoge-nous and exogenous SNAREs. J. Cell Biol. 158:751–760.

22. He, H. T., and D. Marguet. 2011. Detecting nanodomains in living cellmembrane by fluorescence correlation spectroscopy. Annu. Rev. Phys.Chem. 62:417–436.

APP Clustering via the Amyloid-b Domain 1417

23. Pahlsson, P., S. H. Shakin-Eshleman, and S. L. Spitalnik. 1992.N-linked glycosylation of b-amyloid precursor protein. Biochem.Biophys. Res. Commun. 189:1667–1673.

24. Perdivara, I., R. Petrovich, ., M. Przybylski. 2009. Elucidation ofO-glycosylation structures of the b-amyloid precursor protein by liquidchromatography-mass spectrometry using electron transfer dissocia-tion and collision induced dissociation. J. Proteome Res. 8:631–642.

25. Kusumi, A., C. Nakada, ., T. Fujiwara. 2005. Paradigm shift of theplasma membrane concept from the two-dimensional continuum fluidto the partitioned fluid: high-speed single-molecule tracking ofmembrane molecules. Annu. Rev. Biophys. Biomol. Struct. 34:351–378.

26. Rajendran, L., A. Schneider, ., K. Simons. 2008. Efficient inhibitionof the Alzheimer’s disease b-secretase by membrane targeting.Science. 320:520–523.

27. Prox, J., A. Rittger, and P. Saftig. 2011. Physiological functions of theamyloid precursor protein secretases ADAM10, BACE1, and Preseni-lin. Exp Brain Res. 10.1007/s00221-011-2952-0. Epub ahead of printNovember 27, 2011.

28. Munter, L.-M., P. Voigt, ., G. Multhaup. 2007. GxxxG motifs withinthe amyloid precursor protein transmembrane sequence are critical forthe etiology of Ab42. EMBO J. 26:1702–1712.

29. Reinhard, C., S. S. Hebert, and B. De Strooper. 2005. The amyloid-betaprecursor protein: integrating structure with biological function.EMBO J. 24:3996–4006.

30. Beher, D., L. Hesse, ., G. Multhaup. 1996. Regulation of amyloidprotein precursor (APP) binding to collagen and mapping of thebinding sites on APP and collagen type I. J. Biol. Chem. 271:1613–1620.

31. Scheuermann, S., B. Hambsch, ., G. Multhaup. 2001. Homodi-merization of amyloid precursor protein and its implication in theamyloidogenic pathway of Alzheimer’s disease. J. Biol. Chem.276:33923–33929.

32. Wang, Y., and Y. Ha. 2004. The X-ray structure of an antiparalleldimer of the human amyloid precursor protein E2 domain. Mol. Cell.15:343–353.

Biophysical Journal 102(6) 1411–1417