ArticlePDF Available

Gas1 Interferes with A beta PP Trafficking by Facilitating the Accumulation of Immature A beta PP in Endoplasmic Reticulum-Associated Raft Subdomains

Authors:

Abstract and Figures

The amyloid-β protein precursor (AβPP) is a type I transmembrane protein that undergoes maturation during trafficking in the secretory pathway. Proper maturation and trafficking of AβPP are necessary prerequisites for AβPP processing to generate amyloid-β (Aβ), the core component of Alzheimer's disease senile plaques. Recently, we reported that the glycosylphosphatidylinositol (GPI)-anchored protein growth arrest-specific 1 (Gas1) binds to and interferes with the maturation and processing of AβPP. Gas1 expression led to a trafficking blockade of AβPP between the endoplasmic reticulum (ER) and the Golgi. GPI-anchored proteins can exit the ER by transiting through raft subdomains acting as specialized sorting platforms. Here, we show that Gas1 co-partitioned and formed a complex with AβPP in raft fractions, wherein Gas1 overexpression triggered immature AβPP accumulation. Pharmacological interference of ER to Golgi transport increased immature AβPP accumulation upon Gas1 expression in these raft fractions, which were found to be positive for the COPII protein complex component Sec31A, a specific marker for ER exit sites. Furthermore, a Gas1 mutant lacking the GPI anchor that could not transit through rafts was still able to form a complex with AβPP but did not lead to immature AβPP accumulation in rafts. Together these data show that Gas1 interfered with AβPP trafficking by interacting with AβPP to facilitate its translocation into specialized ER-associated rafts where immature AβPP accumulated.
Content may be subject to copyright.
Gas1 Interferes with AβPP Trafficking by Facilitating the
Accumulation of Immature AβPP in Endoplasmic Reticulum-
Associated Raft Subdomains
Julien Chapuisa, Valérie Vingtdeuxa, Hemachander Capirallaa, Peter Daviesa,b, and Philippe
Marambauda,*
aLitwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institute for
Medical Research, Manhasset, NY, USA
bDepartment of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
Abstract
The amyloid-β protein precursor (AβPP) is a type I transmembrane protein that undergoes
maturation during trafficking in the secretory pathway. Proper maturation and trafficking of AβPP
are necessary prerequisites for AβPP processing to generate amyloid-β (Aβ), the core component
of Alzheimer’s disease senile plaques. Recently, we reported that the glycosylphosphatidylinositol
(GPI)-anchored protein growth arrest-specific 1 (Gas1) binds to and interferes with the maturation
and processing of AβPP. Gas1 expression led to a trafficking blockade of AβPP between the
endoplasmic reticulum (ER) and the Golgi. GPI-anchored proteins can exit the ER by transiting
through raft subdomains acting as specialized sorting platforms. Here, we show that Gas1 co-
partitioned and formed a complex with AβPP in raft fractions, wherein Gas1 overexpression
triggered immature AβPP accumulation. Pharmacological interference of ER to Golgi transport
increased immature AβPP accumulation upon Gas1 expression in these raft fractions, which were
found to be positive for the COPII protein complex component Sec31A, a specific marker for ER
exit sites. Furthermore, a Gas1 mutant lacking the GPI anchor that could not transit through rafts
was still able to form a complex with AβPP but did not lead to immature AβPP accumulation in
rafts. Together these data show that Gas1 interfered with AβPP trafficking by interacting with
AβPP to facilitate its translocation into specialized ER-associated rafts where immature AβPP
accumulated.
Keywords
AβPP; Alzheimer’s disease; (Gas1); lipid rafts; protein trafficking
INTRODUCTION
Alzheimer’s disease (AD) is the leading cause of dementia and is due to progressive
neurodegeneration in specific regions of the neocortex and hippocampus [1–3]. Two lesions
are invariably found in the AD brain: the neurofibrillary tangles formed by tau deposition
and senile plaques comprised of aggregated amyloid-β (Aβ) peptides [4, 5]. Aβ is produced
by cleavage of the amyloid-β protein precursor (AβPP) via the sequential action of two
© 2012 – IOS Press and the authors. All rights reserved
*Correspondence to: Philippe Marambaud, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, New
York, NY 11030, USA. Fax: +(516) 562-0401; PMaramba@nshs.edu.
Authors’ disclosures available online (http://www.j-alz.com/disclosures/view.php?id=980).
NIH Public Access
Author Manuscript
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
Published in final edited form as:
J Alzheimers Dis
. 2012 January ; 28(1): 127–135. doi:10.3233/JAD-2011-110434.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
enzymes, β- and γ-secretases [6–9]. The identification of mutations in the AβPP gene
causing familial AD has underscored the central role played by AβPP in AD pathogenesis
[10, 11]. The trafficking of AβPP in the secretory pathway controls AβPP maturation in the
endoplasmic reticulum (ER) and the Golgi before AβPP transport to the cell surface [12,
13]. Proper AβPP trafficking and maturation, via O- and N-glycosylation for instance, are
necessary prerequisites for AβPP accessibility to the secretases at the cell surface or in the
endosomal system and thus for the production of Aβ [14]. In this context, the different
mechanisms controlling AβPP trafficking are under intense investigation.
To identify new genes potentially involved in AD pathogenesis, we previously used a tissue
expression profiling strategy [15, 16] to screen for genes preferentially expressed in the
hippocampus and located in AD chromosomal linkage regions. This approach identified the
CALHM1 (calcium homeostasis modulator 1) and Gas1 (growth arrest-specific 1) genes
[17]. We reported that CALHM1 codes for a novel calcium channel controlling cytosolic
calcium homeostasis and Aβ metabolism [17]. Some studies indicated that CALHM1 may
also control Aβ levels in human cerebrospinal fluid [18, 19] and influence AD age at onset
[17, 20–22]. These results indicated that tissue expression profiling is a valuable approach
for the identification of novel AD candidate genes.
Gas1 is a gene involved in the central nervous system development and in cell cycle control
[23]. Gas1 codes for a glycosylphosphatidylinositol (GPI)-anchored protein required for
normal postnatal proliferation in regions of the brain, such as the cerebellum [24–27]. Gas1
has also been associated with neuronal death in different models of excitotoxicity [28]. GPI-
anchored proteins can exit the ER by transiting through specific coat protein complex II
(COPII)-dependent raft subdomains acting as specialized sorting platforms at ER exit sites
[29, 30]. These ER exit sites express specific protein markers, such as the COPII component
Sec31A, and like plasma membrane lipid rafts, they are characterized by their resistance to
non-ionic detergents. The detergent-resistant property of these subdomains allows their
biochemical isolation in buffers containing Triton X-100 [29, 31].
We reported recently that, although Gas1 did not appear to represent an independent genetic
determinant of AD risk from the available genome-wide association studies, Gas1 can form
a complex with AβPP to control AβPP maturation and trafficking [32]. Gas1 expression was
found to inhibit AβPP full glycosylation and to interfere with AβPP routing to the cell
surface by leading to a trafficking blockade of AβPP between the ER and the Golgi.
Consequently, Gas1 overexpression led to a reduction of Aβ production and conversely,
Gas1 silencing in cells expressing endogenously Gas1 increased Aβ levels [32]. Here, we
show that Gas1 interacted with AβPP in specific raft fractions, wherein Gas1 overexpression
significantly facilitated the accumulation of immature AβPP. Using brefeldin A (BFA)
treatments, we found that interference of ER to Golgi transport increased immature AβPP
accumulation upon Gas1 expression in these raft fractions, which were found to be positive
for the specific ER exit site marker, Sec31A. Furthermore, a GPI-free C-terminally truncated
Gas1 mutant that could not transit through rafts was still able to form a complex with AβPP
but did not lead to immature AβPP accumulation in rafts. Together these data confirm that
Gas1 blocked AβPP trafficking between the ER and the Golgi and further show that Gas1
acted by interacting with AβPP to facilitate its translocation into specialized ER-associated
raft subdomains where immature AβPP accumulated.
MATERIALS AND METHODS
Materials and antibodies
Methyl-β-cyclodextrin (MCD) was purchased from Sigma-Aldrich. BFA was from
Epicentre Technologies. Anti-AβPP-(1–200) LN27 antibody was from Zymed Laboratories
Chapuis et al. Page 2
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
and anti-Aβ-(17–24) 4G8 antibody was from Signet. The anti-AβPP C-terminal domain R1
antibody was provided by Dr. P.D. Mehta (Institute for Basic Research in Developmental
Disabilities, Staten Island, NY). Antibodies directed against Gas1 were purchased from
Santa Cruz Biotechnology (C-17, sc-9585) and Genetex Inc. (GTX101732). Anti-actin, anti-
flotillin-1, and anti-Sec31A antibodies were from BD Transduction Laboratories. Anti-
calreticulin (CRT) antibody was from ABR-Affinity Bio Reagents.
Cell culture, transfections, and western blot (WB) analyses
N2a cells stably transfected with human AβPP695 harboring the Swedish double mutation
(AβPP-N2a) and HEK293 cells stably transfected with human wild type AβPP695 (AβPP-
HEK293) were maintained in 1 : 1 Dulbecco’s modified Eagle’s medium (DMEM)/Opti-
MEM supplemented with 10% fetal bovine serum, penicillin, and streptomycin at 37°C in a
humidified atmosphere with 5% CO2. All cell lines were tested negative for mycoplasma
contaminant [33]. Prior to transfection, cells were plated at a density of ~50%. Transient
transfection (24 h) of Gas1 cDNA (cloned into pCMV6 expression vector, Origene) was
performed using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s
recommendations. For WB, cells were washed with PBS and solubilized in ice-cold lysis
buffer (25 mM HEPES, pH 7.4; 150 mM NaCl; 1% SDS; 1× Complete protease inhibitor
mixture, Roche Applied Sciences). Cell extracts (5–20 μg) were analyzed by SDS-PAGE
using the antibodies listed above. A standard ECL detection procedure was then used.
Sucrose gradient centrifugation
AβPP-HEK293 cells in a 100-mm dish were washed and harvested in ice-cold PBS, and
pelleted by centrifugation for 5 min at 1000 × g at 4°C. Cell pellets were lysed in 1.7 ml of
MBS buffer (25 mM MES, pH 6.5; 150 mM NaCl; 1% CHAPS; 1× Complete protease
inhibitor mixture), disrupted with 10 strokes in a glass Dounce homogenizer with a Teflon
pestle, and sonicated 3 times for 5 s. The resulting cell lysates were mixed with 2.3 ml of
MBS buffer containing 72% sucrose and added to the bottom of ultracentrifuge tubes. MBS
buffer containing 35% sucrose (4 ml) was laid over the cell lysates, followed by 4 ml of
MBS buffer containing 5% sucrose. Samples were centrifuged at 4°C for 16 h at 190,000 ×
g. Twelve fractions of 1 ml each were collected from the top to bottom of the ultracentrifuge
tubes. An aliquot of each fraction was subjected to SDS-PAGE and WB analyses, as
described above.
Preparation of Triton X-100-resistant membranes
Triton X-100-resistant membrane preparations were performed as described before [32].
Briefly, cells were lysed with Triton X-100 lysis buffer (TBS, 1% Triton X-100, 500 μM
sodium orthovanadate, 1× Complete protease inhibitor mixture) and under gentle shaking
for 20 min at 4°C. The crude lysates were then centrifuged at 16,000 × g at 4°C for 10 min.
The supernatants (Triton X-100-soluble fractions) were separated from the pellets and
placed into separate tubes. The pellets were washed with ice-cold Triton X-100 lysis buffer
and centrifuged again. Pellets (Triton X-100-resistant fractions) and Triton X-100-soluble
fractions were then analyzed by WB.
Co-immunoprecipitation assays
Twenty-four hours post-transfection with pCMV6-Gas1 or empty vector, cells were washed
with PBS and Triton X-100-resistant membranes were isolated as described above. Triton
X-100-resistant membranes were resuspended in ice-cold lysis buffer (50 mM Tris-HCl, pH
7.4; 150 mM NaCl; 1 mM EDTA; 0.25% deoxycholic acid; 1% NP-40; 1% Triton X-100; 1
× Complete protease inhibitor mixture) and disrupted with 10 strokes in a glass Dounce
homogenizer with a Teflon pestle and centrifuged at 16,000 × g for 5 min at 4°C. The
Chapuis et al. Page 3
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
supernatants were pre-cleared with 50 μl protein-G for 2 h at 4°C. After centrifugation at
1,500 × g for 30 s, supernatants were incubated overnight at 4°C with the indicated
antibodies. The following day, antibodies were precipitated by incubation with protein-G for
2 h at 4°C. Precipitates were washed 3 times with lysis buffer and then were resuspended in
loading buffer (50 mM Tris, pH 6.8; 5% β-mercaptoethanol; 2% SDS; 0.1% bromophenol
blue; 10% glycerol) and resolved by SDS-PAGE.
Statistical analysis
The results are presented as the mean ± standard deviation (S.D.). Significant differences
between groups were determined using the unpaired Student’s t-test. For all analyses, p <
0.05 was considered statistically significant.
RESULTS
Gas1 induces the accumulation of immature AβPP in rafts
Recently, we reported that Gas1 inhibited AβPP maturation and processing by leading to a
trafficking blockade of AβPP between the ER and the Golgi [32]. Because Gas1, like many
other GPI-anchored proteins, is known to localize in rafts, we asked whether Gas1
expression controls AβPP levels in rafts. Using sucrose gradient centrifugation, we observed
a significant shift of AβPP into the flotillin-positive raft fraction in Gas1-transfected cells, as
compared to control cells (Fig. 1A, fraction 5, and Fig. 1B). The effect of Gas1 expression
on AβPP translocation into rafts was confirmed by another method using Triton X-100-
resistant membrane preparations [34]. Similar to the observation using sucrose gradient
centrifugation, Gas1 expression resulted in a robust accumulation of AβPP in flotillin-
positive detergent-resistant raft fractions (Fig. 1C). Protein analysis using higher resolution
WB showed that low-molecular-weight immature AβPP specifically accumulated in raft
fractions upon Gas1 expression, whereas mature AβPP and AβPP C-terminal fragments,
CTFα and CTFβ, were detected at low levels in these fractions and were not significantly
affected by Gas1 expression (Fig. 1D and E). Of note, Gas1 interfered with AβPP trafficking
both in cells expressing wild type (AβPP-HEK293, Fig. 1A) and Swedish (AβPP-N2a, Fig.
1C) AβPP, suggesting that the Swedish mutation on AβPP did not impact Gas1 control of
AβPP trafficking. Rafts and detergent-resistant membrane fractions are enriched in
cholesterol and sphingolipids, two critical structural components for raft formation.
Cholesterol depletion using methyl-β-cyclodextrin was found to lower the accumulation of
both Gas1 and immature AβPP in detergent-resistant fractions (Fig. 1F and G), confirming
that immature AβPP accumulated and co-partitioned with Gas1 in rafts in Gas1-transfected
cells.
Gas1 and AβPP form a complex in rafts
Co-immunoprecipitation experiments previously revealed that Gas1 and AβPP can form a
complex in cells [32]. Here, we asked whether this interaction takes place in rafts by
performing reciprocal immunoprecipitations with antibodies directed against Gas1 or AβPP
in detergent-resistant raft fractions resuspended in lysis buffer containing raft disrupting
detergents. We found that Gas1 co-immunoprecipitated with AβPP in cells expressing the
two proteins (Fig. 2A). These results indicated that Gas1/AβPP interaction can occur in
rafts. We also observed that Gas1 preferentially co-precipitated with immature AβPP,
suggesting that Gas1 interacted with AβPP early in the secretory pathway prior to AβPP
maturation.
To confirm that Gas1 controls AβPP translocation into rafts, we asked whether inhibition of
Gas1 trafficking in rafts affects AβPP accumulation in rafts upon Gas1 expression. Because
incorporation of GPI-anchored proteins into rafts is selectively driven by their GPI anchor, a
Chapuis et al. Page 4
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Gas1 construct was generated in which the GPI consensus sequence was removed (Fig. 2B).
We confirmed the inability of this GPI-free soluble Gas1 mutant (sGas1) to translocate into
rafts (Fig. 2C). We found that GPI-free sGas1 was still able to form a complex with AβPP
(Fig. 2D) but failed to trigger the accumulation of immature AβPP in rafts (Fig. 2E),
showing that Gas1 translocation into rafts is required for its effect on AβPP accumulation in
rafts. These results further demonstrate that the GPI anchor is dispensable for the Gas1/
AβPP interaction.
Gas1 controls immature AβPP translocation into ER-associated rafts
Gas1 expression led to the accumulation of immature AβPP in rafts (Figs. 1 and 2),
suggesting that the two proteins interacted early in the secretory pathway in a compartment
close to the ER. Because GPI-anchored proteins can exit the ER by transiting through
specific ER sorting platforms [29, 30], we next asked whether immature AβPP accumulates
and co-partitions with Gas1 in ER-associated rafts. We previously reported that while the
Triton X-100-soluble fractions are immunoreactive for calreticulin and therefore contained
most of the ER, the Triton X-100-resistant fractions were found to be strongly
immunoreactive for Sec31A and thus are enriched in ER exit sites, indicating that Gas1 may
facilitate AβPP accumulation at these specific ER-associated subdomains [32]. To go
further, we investigated whether interference of ER to Golgi transport prevents the effect of
Gas1 expression on AβPP translocation into rafts. Treatments with BFA, an agent that
blocks protein transport from the ER to the Golgi by disassembling the Golgi complex [35],
failed to inhibit immature AβPP accumulation in raft fractions, which were found to be still
positive for the ER exit site marker Sec31A and negative for the ER marker calreticulin
(Fig. 3A and B). Instead, we found that BFA treatments slightly but significantly
strengthened the effect of Gas1 expression on immature AβPP accumulation in rafts (Fig.
3A and B), confirming that AβPP accumulated in a compartment localized between the ER
and the Golgi. Altogether these data show that Gas1 expression led to immature AβPP
accumulation in ER-associated rafts.
DISCUSSION
Strong evidence indicates that lipid rafts are involved in AβPP amyloidogenic processing
[36, 37]. Lipid rafts are cholesterol- and sphingolipid-rich membrane microdomains and
studies in cell cultures and animal models have shown that alteration in cholesterol and
sphingolipid distribution or metabolism can significantly impact AβPP processing and Aβ
production [37, 38]. Both BACE1/β-secretase and γ-secretase are enriched in lipid rafts and
can target AβPP for Aβ production in these microdomains. γ-Secretase is involved in
regulated intramembranous proteolysis of multiple type I transmembrane proteins beyond
AβPP, such as Notch or N- and E-cadherins [8, 39]. γ-Secretase in the adult brain, however,
appears to preferentially target AβPP in lipid rafts [40]. Together, these results motivate
further studies aimed at determining whether targeting raft-dependent amyloidogenic
processing of AβPP is therapeutically relevant.
Lipid raft biogenesis and cellular trafficking is complex. Most rafts are found at the plasma
membrane where they follow the endocytotic pathway to recycle back to the plasma
membrane or to return to the Golgi. Rafts found at the plasma membrane are mostly
generated at the Golgi and use the anterograde pathway to traffic to the plasma membrane
[41–43]. Some rafts, however, can be generated from the ER membrane and GPI-anchored
proteins can use these rafts to exit the ER. These specific ER rafts are COPII-dependent
subdomains acting as specialized sorting platforms [29, 30, 44]. Like plasma membrane
lipid rafts, rafts at ER exit sites are characterized by their resistance to Triton X-100 at 4°C
[29, 31]. At steady state, it is estimated that about 10% of AβPP is targeted to lipid rafts and
that raft-localized AβPP is mostly regulated by endocytosis [45]. Here, we reveal another
Chapuis et al. Page 5
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
level of complexity in AβPP trafficking in rafts. Using Triton X-100-resistant membrane
preparations, we observed that Gas1 physically interacted with AβPP to co-partition in rafts
at ER exit sites. It is important to note that the use of non-ionic detergents to isolate rafts and
study protein trafficking in these microdomains has raised some concerns [46]. In this
context, we also used a GPI-free Gas1 mutant that lost its properties to traffic through rafts
to show that Gas1 translocation into rafts was required for its effect on AβPP accumulation
in rafts. Thus, the current work shows that AβPP can also accumulate in specialized ER-
associated rafts. Although further studies will be required to determine the relative amounts
at steady state of AβPP transiting through these rafts and what might be the contribution of
this pathway to constitutive AβPP processing, this work suggests that approaches aimed at
increasing AβPP translocation in these ER-associated subdomains (e.g., by increasing Gas1
expression) can significantly influence AβPP maturation and processing, and thus Aβ
production [32].
In summary, we provide strong evidence that Gas1 negatively controlled AβPP maturation
by inhibiting AβPP trafficking between the ER and the Golgi and this by physically
interacting with AβPP to trigger its translocation into specialized ER-associated raft
subdomains where immature AβPP accumulated. This study reveals a new facet of AβPP
trafficking and further increases our understanding of Gas1 function.
Acknowledgments
We thank Dr. G. Thinakaran (University of Chicago, Chicago, IL, USA) for kindly providing us with AβPP-N2a
cells, Dr. L. D’Adamio (Albert Einstein College of Medicine, Bronx, NY, USA) for AβPP-HEK293 cells; Dr P.D.
Mehta (Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA) for anti-AβPP R1
antibody. J.C. is the recipient of a fellowship from the Philippe Foundation (New York, NY, USA).
References
1. Selkoe DJ. Alzheimer’s disease: Genes, proteins, and therapy. Physiol Rev. 2001; 81:741–766.
[PubMed: 11274343]
2. Citron M. Alzheimer’s disease: Strategies for disease modification. Nat Rev Drug Discov. 2010;
9:387–398. [PubMed: 20431570]
3. Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010; 362:329–344. [PubMed:
20107219]
4. Duyckaerts C, Delatour B, Potier MC. Classification and basic pathology of Alzheimer disease.
Acta Neuropathol. 2009; 118:5–36. [PubMed: 19381658]
5. Buee L, Bussiere T, Buee-Scherrer V, Delacourte A, Hof PR. Tau protein isoforms, phosphorylation
and role in neurodegenerative disorders. Brain Res Brain Res Rev. 2000; 33:95–130. [PubMed:
10967355]
6. Checler F. Processing of the beta-amyloid precursor protein and its regulation in Alzheimer’s
disease. J Neurochem. 1995; 65:1431–1444. [PubMed: 7561836]
7. Wilquet V, De Strooper B. Amyloid-beta precursor protein processing in neurodegeneration. Curr
Opin Neurobiol. 2004; 14:582–588. [PubMed: 15464891]
8. Marambaud P, Robakis NK. Genetic and molecular aspects of Alzheimer’s disease shed light on
new mechanisms of transcriptional regulation. Genes Brain Behav. 2005; 4:134–146. [PubMed:
15810902]
9. Sambamurti K, Greig NH, Lahiri DK. Advances in the cellular and molecular biology of the beta-
amyloid protein in Alzheimer’s disease. Neuromolecular Med. 2002; 1:1–31. [PubMed: 12025813]
10. Ertekin-Taner N. Genetics of Alzheimer disease in the pre- and post-GWAS era. Alzheimers Res
Ther. 2010; 2:3. [PubMed: 20236449]
11. Lambert JC, Amouyel P. Genetic heterogeneity of Alzheimer’s disease: Complexity and advances.
Psychoneuroendocrinology. 2007; 32(Suppl 1):S62–S70. [PubMed: 17659844]
Chapuis et al. Page 6
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
12. Thinakaran G, Koo EH. Amyloid precursor protein trafficking, processing, and function. J Biol
Chem. 2008; 283:29615–29619. [PubMed: 18650430]
13. Suzuki T, Araki Y, Yamamoto T, Nakaya T. Trafficking of Alzheimer’s disease-related membrane
proteins and its participation in disease pathogenesis. J Biochem. 2006; 139:949–955. [PubMed:
16788045]
14. Sannerud R, Annaert W. Trafficking, a key player in regulated intramembrane proteolysis. Semin
Cell Dev Biol. 2009; 20:183–190. [PubMed: 19056506]
15. Skrabanek L, Campagne F. TissueInfo: High-throughput identification of tissue expression profiles
and specificity. Nucleic Acids Res. 2001; 29:E102. [PubMed: 11691939]
16. Aguilar D, Skrabanek L, Gross SS, Oliva B, Campagne F. Beyond tissueInfo: Functional
prediction using tissue expression profile similarity searches. Nucleic Acids Res. 2008; 36:3728–
3737. [PubMed: 18483083]
17. Dreses-Werringloer U, Lambert JC, Vingtdeux V, Zhao H, Vais H, Siebert A, Jain A, Koppel J,
Rovelet-Lecrux A, Hannequin D, Pasquier F, Galimberti D, Scarpini E, Mann D, Lendon C,
Campion D, Amouyel P, Davies P, Foskett JK, Campagne F, Marambaud P. A polymorphism in
CALHM1 influences Ca2+ homeostasis, Abeta levels, and Alzheimer’s disease risk. Cell. 2008;
133:1149–1161. [PubMed: 18585350]
18. Kauwe JS, Cruchaga C, Bertelsen S, Mayo K, Latu W, Nowotny P, Hinrichs AL, Fagan AM,
Holtzman DM, Goate AM. Alzheimer’s Disease Neuroimaging Initiative. Validating predicted
biological effects of Alzheimer’s disease associated SNPs using CSF biomarker levels. J
Alzheimers Dis. 2010; 21:833–842. [PubMed: 20634593]
19. Koppel J, Campagne F, Vingtdeux V, Dreses-Werringloer U, Ewers M, Rujescu D, Hampel H,
Gordon ML, Christen E, Chapuis J, Greenwald BS, Davies P, Marambaud P. CALHM1 P86L
polymorphism modulates CSF Abeta levels in cognitively healthy individuals at risk for
Alzheimer’s disease. Mol Med. 201110.2119/molmed.2011.00154
20. Boada M, Antunez C, Lopez-Arrieta J, Galan JJ, Moron FJ, Hernandez I, Marin J, Martinez-Lage
P, Alegret M, Carrasco JM, Moreno C, Real LM, Gonzalez-Perez A, Tarraga L, Ruiz A.
CALHM1 P86L polymorphism is associated with late-onset Alzheimer’s disease in a recessive
model. J Alzheimers Dis. 2010; 20:247–251. [PubMed: 20164592]
21. Minster RL, Demirci FY, DeKosky ST, Kamboh MI. No association between CALHM1 variation
and risk of Alzheimer disease. Hum Mutat. 2009; 30:E566–E569. [PubMed: 19191331]
22. Lambert JC, Sleegers K, Gonzalez-Perez A, Ingelsson M, Beecham GW, Hiltunen M, Combarros
O, Bullido MJ, Brouwers N, Bettens K, Berr C, Pasquier F, Richard F, Dekosky ST, Hannequin D,
Haines JL, Tognoni G, Fievet N, Dartigues JF, Tzourio C, Engelborghs S, Arosio B, Coto E, De
Deyn P, Del Zompo M, Mateo I, Boada M, Antunez C, Lopez-Arrieta J, Epelbaum J, Schjeide
BM, Frank-Garcia A, Giedraitis V, Helisalmi S, Porcellini E, Pilotto A, Forti P, Ferri R, Delepine
M, Zelenika D, Lathrop M, Scarpini E, Siciliano G, Solfrizzi V, Sorbi S, Spalletta G, Ravaglia G,
Valdivieso F, Vepsalainen S, Alvarez V, Bosco P, Mancuso M, Panza F, Nacmias B, Bossu P,
Hanon O, Piccardi P, Annoni G, Mann D, Marambaud P, Seripa D, Galimberti D, Tanzi RE,
Bertram L, Lendon C, Lannfelt L, Licastro F, Campion D, Pericak-Vance MA, Soininen H, Van
Broeckhoven C, Alperovitch A, Ruiz A, Kamboh MI, Amouyel P. The CALHM1 P86L
polymorphism is a genetic modifier of age at onset in Alzheimer’s disease: A meta-analysis study.
J Alzheimers Dis. 2010; 22:247–255. [PubMed: 20847397]
23. Martinelli DC, Fan CM. The role of Gas1 in embryonic development and its implications for
human disease. Cell Cycle. 2007; 6:2650–2655. [PubMed: 17726382]
24. Schueler-Furman O, Glick E, Segovia J, Linial M. Is GAS1 a co-receptor for the GDNF family of
ligands? Trends Pharmacol Sci. 2006; 27:72–77. [PubMed: 16406089]
25. Cabrera JR, Sanchez-Pulido L, Rojas AM, Valencia A, Manes S, Naranjo JR, Mellstrom B. Gas1 is
related to the glial cell-derived neurotrophic factor family receptors alpha and regulates Ret
signaling. J Biol Chem. 2006; 281:14330–14339. [PubMed: 16551639]
26. Marques G, Fan CM. Growth arrest specific gene 1: A fuel for driving growth in the cerebellum.
Cerebellum. 2002; 1:259–263. [PubMed: 12879964]
27. Kang JS, Zhang W, Krauss RS. Hedgehog signaling: Cooking with Gas1. Sci STKE. 2007;
2007:pe50. [PubMed: 17848687]
Chapuis et al. Page 7
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
28. Mellstrom B, Cena V, Lamas M, Perales C, Gonzalez C, Naranjo JR. Gas1 is induced during and
participates in excitotoxic neuronal death. Mol Cell Neurosci. 2002; 19:417–429. [PubMed:
11906213]
29. Bonnon C, Wendeler MW, Paccaud JP, Hauri HP. Selective export of human GPI-anchored
proteins from the endoplasmic reticulum. J Cell Sci. 2010; 123:1705–1715. [PubMed: 20427317]
30. Mayor S, Riezman H. Sorting GPI-anchored proteins. Nat Rev Mol Cell Biol. 2004; 5:110–120.
[PubMed: 15040444]
31. Sevlever D, Pickett S, Mann KJ, Sambamurti K, Medof ME, Rosenberry TL.
Glycosylphosphatidylinositol-anchor intermediates associate with triton-insoluble membranes in
subcellular compartments that include the endoplasmic reticulum. Biochem J. 1999; 343 (Pt 3):
627–635. [PubMed: 10527942]
32. Chapuis J, Vingtdeux V, Campagne F, Davies P, Marambaud P. Growth arrest-specific 1 binds to
and controls the maturation and processing of the amyloid-beta precursor protein. Hum Mol
Genet. 2011; 20:2026–2036. [PubMed: 21357679]
33. Zhao H, Dreses-Werringloer U, Davies P, Marambaud P. Amyloid-beta peptide degradation in cell
cultures by mycoplasma contaminants. BMC Res Note. 2008; 1:38.
34. Pierchala BA, Milbrandt J, Johnson EM Jr. Glial cell line-derived neurotrophic factor-dependent
recruitment of Ret into lipid rafts enhances signaling by partitioning Ret from proteasome-
dependent degradation. J Neurosci. 2006; 26:2777–2787. [PubMed: 16525057]
35. Klausner RD, Donaldson JG, Lippincott-Schwartz J. Brefeldin A: Insights into the control of
membrane traffic and organelle structure. J Cell Biol. 1992; 116:1071–1080. [PubMed: 1740466]
36. Rushworth JV, Hooper NM. Lipid Rafts: Linking Alzheimer’s Amyloid-beta production,
aggregation, and toxicity at neuronal membranes. Int J Alzheimers Dis. 2010; 2011:603052.
[PubMed: 21234417]
37. Vetrivel KS, Thinakaran G. Membrane rafts in Alzheimer’s disease beta-amyloid production.
Biochim Biophys Acta. 2010; 1801:860–867. [PubMed: 20303415]
38. Sawamura N, Ko M, Yu W, Zou K, Hanada K, Suzuki T, Gong JS, Yanagisawa K, Michikawa M.
Modulation of amyloid precursor protein cleavage by cellular sphingolipids. J Biol Chem. 2004;
279:11984–11991. [PubMed: 14715666]
39. Lichtenthaler SF, Haass C, Steiner H. Regulated intramembrane proteolysis–lessons from amyloid
precursor protein processing. J Neurochem. 2011; 117:779–796. [PubMed: 21413990]
40. Vetrivel KS, Cheng H, Kim SH, Chen Y, Barnes NY, Parent AT, Sisodia SS, Thinakaran G.
Spatial segregation of gamma-secretase and substrates in distinct membrane domains. J Biol
Chem. 2005; 280:25892–25900. [PubMed: 15886206]
41. Brugger B, Sandhoff R, Wegehingel S, Gorgas K, Malsam J, Helms JB, Lehmann WD, Nickel W,
Wieland FT. Evidence for segregation of sphingomyelin and cholesterol during formation of
COPI-coated vesicles. J Cell Biol. 2000; 151:507–518. [PubMed: 11062253]
42. Brown DA, London E. Functions of lipid rafts in biological membranes. Annu Rev Cell Dev Biol.
1998; 14:111–136. [PubMed: 9891780]
43. Helms JB, Zurzolo C. Lipids as targeting signals: Lipid rafts and intracellular trafficking. Traffic.
2004; 5:247–254. [PubMed: 15030566]
44. Glick BS. ER export: More than one way out. Curr Biol. 2001; 11:R361–R363. [PubMed:
11369248]
45. Ehehalt R, Keller P, Haass C, Thiele C, Simons K. Amyloidogenic processing of the Alzheimer
beta-amyloid precursor protein depends on lipid rafts. J Cell Biol. 2003; 160:113–123. [PubMed:
12515826]
46. Lichtenberg D, Goni FM, Heerklotz H. Detergent-resistant membranes should not be identified
with membrane rafts. Trends Biochem Sci. 2005; 30:430–436. [PubMed: 15996869]
Chapuis et al. Page 8
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 1.
Gas1 induces the accumulation of immature AβPP in rafts. A) AβPP-HEK293 cells
transiently transfected with Gas1 cDNA or empty vector were lysed in 1% CHAPS buffer
and subjected to discontinuous sucrose density gradient ultracentrifugation fractionation. All
fractions (1–12) were analyzed by WB using antibodies directed against AβPP, Gas1,
calreticulin (CRT, ER resident protein), and flotillin-1 (lipid raft resident protein). B) Shown
are densitometric analysis and quantification of AβPP immunoreactivity in fraction 5 (raft
AβPP) in three independent experiments performed as in A. C) AβPP-N2a transiently
transfected with Gas1 cDNA (+) or empty vector () were lysed in 1% Triton X-100 buffer
at 4°C and Triton X-100-soluble and Triton X-100-resistant fractions (see Materials and
Methods) were analyzed by WB for the indicated proteins. D) WB analysis of protein
extracts from cells treated as in C showing the accumulation of immature AβPP in Triton
X-100-resistant fractions upon Gas1 transfection. ma. AβPP, mature AβPP; im. AβPP,
immature AβPP. E) Shown are densitometric analysis and quantification of immature and
mature AβPP levels in three independent experiments performed as in D. F) AβPP-HEK293
cells transiently transfected with Gas1 cDNA (+) or empty vector () were treated (+) or not
() with methyl-β-cyclodextrin (MCD, 10 mM, 1 h). Triton X-100-soluble and Triton
Chapuis et al. Page 9
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
X-100-resistant fractions were then analyzed by WB using antibodies directed against the
indicated proteins. Note that the effect of MCD on raft integrity was confirmed by the partial
translocation of flotillin in the Triton X-100-soluble fractions (3rd and 4th lanes). G) Shown
are densitometric analysis and quantification of immature AβPP levels in Triton X-100-
resistant fractions in three independent experiments performed as in F. Histograms in B, E,
and G indicate the mean ± S.D. a.u., arbitrary units. *p < 0.05, **p < 0.01 (Student’s t-test).
Chapuis et al. Page 10
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 2.
Gas1 and AβPP form a complex in rafts. A) Triton X-100 resistant fraction homogenates
from AβPP-HEK293 cells transiently transfected with Gas1 cDNA (+) or empty vector ()
were incubated in the absence (CTRL) or presence of anti-AβPP (4G8) or anti-Gas1 (C-17)
antibodies. Total extracts from Triton X-100 resistant fractions (Input) and
immunoprecipitated (IP) proteins were analyzed by WB using antibodies directed against
AβPP (LN27) and Gas1 (GTX101732). B) Map of the constructs generated to express Gas1
full-length and soluble Gas1 (sGas1) lacking the GPI consensus sequence. C) AβPP-N2a
cells transiently transfected (+) or not () with Gas1 or sGas1 cDNAs were lysed in 1%
Triton X-100 buffer at 4°C and Triton X-100-soluble and Triton X-100-resistant fractions
were analyzed by WB for the indicated proteins. D) AβPP-HEK293 cells transiently
transfected with sGas1 cDNA (+) or empty vector () were IP with anti-AβPP (4G8)
antibody. Total extract (Input) and precipitated proteins were analyzed by WB using
antibodies directed against AβPP (LN27), Gas1 (GTX101732), and actin. E) AβPP-HEK293
cells transiently transfected with empty vector, Gas1 cDNA, or sGas1 cDNA were lysed in
1% Triton X-100 buffer at 4°C. Triton X-100-soluble and Triton X-100-resistant fractions
were then analyzed by WB for the indicated proteins.
Chapuis et al. Page 11
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 3.
Gas1 controls immature AβPP translocation into ER-associated rafts. A) AβPP-N2a cells
transiently transfected with Gas1 (+) or empty vector () were treated with brefeldin A
(BFA, 5 μg/ml) for the indicted times. Cells were then lysed in 1% Triton X-100 buffer at
4°C and Triton X-100-soluble and Triton X-100-resistant fractions were analyzed by WB
for the indicated proteins. B) Shown are densitometric analysis and quantification of
immature and mature AβPP levels in three independent experiments performed as in A.
Histogram indicates the mean ± S.D. a.u., arbitrary units. *p < 0.05, **p < 0.01 (Student’s t-
test).
Chapuis et al. Page 12
J Alzheimers Dis. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... In addition, Gas1 controls the maturation and processing of the β-amyloid precursor protein (APP) and thus, the production of amyloidbeta (Aβ), the core component of the Alzheimer's disease senile plaques. (Chapuis et al. 2011(Chapuis et al. , 2012. In the embryo mouse brain, Gas1 is expressed in the diencephalon, midbrain, and hindbrain, and particularly in progenitor cells of the cortex and dentate gyrus Lee and Fan 2001). ...
... In addition, we found that the levels of Gas1 increased gradually as the treatment with RA progressed (Fig. 8a, b). These data are consistent with a previous report (Chapuis et al. 2012). On the other hand, RA promotes neuronal differentiation in SH-SY5Y cells, inducing a predominantly mature dopaminegic-like phenotype. ...
... 5). On the other hand, Gas1 regulates the maturation and reduces the levels of amyloid-β precursor protein (APP) in SH-SY5Y cells (Chapuis et al. 2012), suggesting that Gas1 might have a relevant role in Alzheimer's disease. ...
Article
Full-text available
Growth arrest-specific 1 (Gas1) is a pleiotropic protein that induces apoptosis of tumor cells and has important roles during development. Recently, the presence of two forms of Gas1 was reported: one attached to the cell membrane by a GPI anchor; and a soluble extracellular form shed by cells. Previously, we showed that Gas1 is expressed in different areas of the adult mouse CNS. Here, we report the levels of Gas1 mRNA protein in different regions and analyzed its expressions in glutamatergic, GABAergic, and dopaminergic neurons. We found that Gas1 is expressed in GABAergic and glutamatergic neurons in the Purkinje-molecular layer of the cerebellum, hippocampus, thalamus, and fastigial nucleus, as well as in dopaminergic neurons of the substantia nigra. In all cases, Gas1 was found in the cell bodies, but not in the neuropil. The Purkinje and the molecular layers show the highest levels of Gas1, whereas the granule cell layer has low levels. Moreover, we detected the expression and release of Gas1 from primary cultures of Purkinje cells and from hippocampal neurons as well as from neuronal cell lines, but not from cerebellar granular cells. In addition, using SH-SY5Y cells differentiated with retinoic acid as a neuronal model, we found that extracellular Gas1 promotes neurite outgrowth, increases the levels of tyrosine hydroxylase, and stimulates the inhibition of GSK3β. These findings demonstrate that Gas1 is expressed and released by neurons and promotes differentiation, suggesting an important role for Gas1 in cellular signaling in the CNS.
... These data indicate that GAS1 is a pleiotropic molecule whose functions differ depending on the cellular context. To support this statement, recent reports associate GAS1 with muscle differentiation (Leem et al. 2011), heart development (Xie et al. 2012), the processing of ß-amyloid (Chapuis et al. 2011;Chapuis et al. 2012), and the proliferation of mesangial cells in the kidney (vanRoeyen et al. 2013). Considering the previous information, particularly the expression of GAS1 during development, it becomes relevant to determine whether GAS1 is present and exerts a function in the adult brain. ...
... GAS1 is a GPI-linked protein, and this suggests that it may exert effects by binding with other signaling regulators including Ret, SHH, or GFRα1 and, in this manner, contribute to brain maintenance. Recently, it was demonstrated that GAS1 forms a complex with APP thatnegatively controls the vesicular trafficking of APP from the ER to the Golgi, and thus its maturation and processing (Chapuis et al. 2012). Here, we show that GAS1 is abundantly expressed in neurons, with a particularly high expression in hippocampus; hence, it will be relevant to further investigate this activity in the contexts of normal brain physiology and of its potential participation in Alzheimer's disease. ...
Article
Full-text available
Growth arrest specific 1 (GAS1) is a pleiotropic protein that induces apoptosis and cell arrest in different tumors, but it is also involved in the development of the nervous system and other tissues and organs. This dual ability is likely caused by its capacity to interact both by inhibiting the intracellular signaling cascade induced by glial cell-line derived neurotrophic factor and by facilitating the activity of the sonic hedgehog pathway. The presence of GAS1 mRNA has been described in adult mouse brain, and here we corroborated this observation. We then proceeded to determine the distribution of the protein in the adult central nervous system (CNS). We detected, by western blot analysis, expression of GAS1 in olfactory bulb, caudate-putamen, cerebral cortex, hippocampus, mesencephalon, medulla oblongata, cerebellum, and cervical spinal cord. To more carefully map the expression of GAS1, we performed double-label immunohistochemistry and noticed expression of GAS1 in neurons in all brain areas examined. We also observed expression of GAS1 in astroglial cells, albeit the pattern of expression was more restricted than that seen in neurons. Briefly, in the present article, we report the widespread distribution and cellular localization of the GAS1 native protein in adult mammalian CNS.
... Gas1 binds to the Amyloid precursor protein (APP) and inhibits its glycosylation, as well as its transport to the cellular surface regulating the transit of APP from the ER to the Golgi. In summary Gas1 regulates the traffic, processing and maturation of APP which may imply a reduction in the Aβ formation [97,98]; however more research is needed to take advantage of this capacity of Gas1 as a therapeutic strategy in Alzheimer´s Disease (AD). ...
Article
Cellular homeostasis is governed by a precise regulation of the molecular mechanisms of action of several proteins in a given time. There is a group of proteins that have a particular role depending on the cellular context in which they are present and are known as pleiotropic proteins. The Gas1 (Growth Arrest Specific 1) gene was isolated from a subtraction library from serum arrested versus growing NIH-3T3 mouse fibroblast. Gas1 is a member of the alpha receptors (GFRα) for the family of GDNF ligands (GFL), we have previously shown that Gas1 acts as a negative modulator of the GDNF-induced intracellular signaling and induces cell arrest and apoptosis. This modulating activity is the cause of the capacity of Gas1 to act as a tumor suppressor. On the other hand, several studies have shown the interaction between Gas1 and Hh (Hedgehog) proteins to potentiate the positive regulation of this pathway, which is involved in the development of the nervous system, and in both the origin and progression of different tumors. This review summarizes our current understanding of the structure of Gas1 and the molecular mechanism of action in different cellular functions, both during embryonic development, in the adult and its effects inhibiting cell growth and inducing apoptosis of cancer cells.
Article
Full-text available
In higher eukaryotes, phospholipid and cholesterol synthesis occurs mainly in the endoplasmic reticulum, whereas sphingomyelin and higher glycosphingolipids are synthesized in the Golgi apparatus. Lipids like cholesterol and sphingomyelin are gradually enriched along the secretory pathway, with their highest concentration at the plasma membrane. How a cell succeeds in maintaining organelle-specific lipid compositions, despite a steady flow of incoming and outgoing transport carriers along the secretory pathway, is not yet clear. Transport and sorting along the secretory pathway of both proteins and most lipids are thought to be mediated by vesicular transport, with coat protein I (COPI) vesicles operating in the early secretory pathway. Although the protein constituents of these transport intermediates are characterized in great detail, much less is known about their lipid content. Using nano-electrospray ionization tandem mass spectrometry for quantitative lipid analysis of COPI-coated vesicles and their parental Golgi membranes, we find only low amounts of sphingomyelin and cholesterol in COPI-coated vesicles compared with their donor Golgi membranes, providing evidence for a significant segregation from COPI vesicles of these lipids. In addition, our data indicate a sorting of individual sphingomyelin molecular species. The possible molecular mechanisms underlying this segregation, as well as implications on COPI function, are discussed.
Article
Full-text available
Intracellular trafficking and proteolytic processing of amyloid precursor protein (APP) have been the focus of numerous investigations over the past two decades. APP is the precursor to the amyloid β-protein (Aβ), the 38–43-amino acid residue peptide that is at the heart of the amyloid cascade hypothesis of Alzheimer disease (AD). Tremendous progress has been made since the initial identification of Aβ as the principal component of brain senile plaques of individuals with AD. Specifically, molecular characterization of the secretases involved in Aβ production has facilitated cell biological investigations on APP processing and advanced efforts to model AD pathogenesis in animal models. This minireview summarizes salient features of APP trafficking and amyloidogenic processing and discusses the putative biological functions of APP.
Article
Full-text available
The calcium homeostasis modulator 1 (CALHM1) gene codes for a novel cerebral calcium channel controlling intracellular calcium homeostasis and amyloid-β (Aβ) peptide metabolism, a key event in the etiology of Alzheimer's disease (AD). The P86L polymorphism in CALHM1 (rs2986017) initially was proposed to impair CALHM1 functionally and to lead to an increase in Aβ accumulation in vitro in cell lines. Recently, it was reported that CALHM1 P86L also may influence Aβ metabolism in vivo by increasing Aβ levels in human cerebrospinal fluid (CSF). Although the role of CALHM1 in AD risk remains uncertain, concordant data have now emerged showing that CALHM1 P86L is associated with an earlier age at onset of AD. Here, we have analyzed the association of CALHM1 P86L with CSF Aβ in samples from 203 AD cases and 46 young cognitively healthy individuals with a positive family history of AD. We failed to detect an association between the CALHM1 polymorphism and CSF Aβ levels in AD patients. Our data, however, revealed a significant association of CALHM1 P86L with elevated CSF Aβ42 and Aβ40 in the normal cohort at risk for AD. This work shows that CALHM1 modulates CSF Aβ levels in presymptomatic individuals, strengthening the notion that CALHM1 is involved in AD pathogenesis. These data further demonstrate the utility of endophenotype-based approaches focusing on CSF biomarkers for the identification or validation of risk factors for AD.
Article
Full-text available
Alzheimer's disease (AD), the most common neurodegenerative disorder, is characterized by cerebral deposition of amyloid-β (Aβ), a series of peptides derived from the processing of the amyloid-β precursor protein (APP). To identify new candidate genes for AD, we recently performed a transcriptome analysis to screen for genes preferentially expressed in the hippocampus and located in AD linkage regions. This strategy identified CALHM1 (calcium homeostasis modulator 1), a gene modulating AD age at onset and Aβ metabolism. Here, we focused our attention on another candidate identified using this screen, growth arrest-specific 1 (Gas1), a gene involved in the central nervous system development. We found that Gas1 formed a complex with APP and controlled APP maturation and processing. Gas1 expression inhibited APP full glycosylation and routing to the cell surface by leading to a trafficking blockade of APP between the endoplasmic reticulum and the Golgi. Gas1 expression also resulted in a robust inhibition of APP transport into multivesicular bodies, further demonstrating that Gas1 negatively regulated APP intracellular trafficking. Consequently, Gas1 overexpression led to a reduction in Aβ production, and conversely, Gas1 silencing in cells expressing endogenously Gas1 increased Aβ levels. These results suggest that Gas1 is a novel APP-interacting protein involved in the control of APP maturation and processing.
Article
Full-text available
Lipid rafts are membrane microdomains, enriched in cholesterol and sphingolipids, into which specific subsets of proteins and lipids partition, creating cell-signalling platforms that are vital for neuronal functions. Lipid rafts play at least three crucial roles in Alzheimer's Disease (AD), namely, in promoting the generation of the amyloid-β (Aβ) peptide, facilitating its aggregation upon neuronal membranes to form toxic oligomers and hosting specific neuronal receptors through which the AD-related neurotoxicity and memory impairments of the Aβ oligomers are transduced. Recent evidence suggests that Aβ oligomers may exert their deleterious effects through binding to, and causing the aberrant clustering of, lipid raft proteins including the cellular prion protein and glutamate receptors. The formation of these pathogenic lipid raft-based platforms may be critical for the toxic signalling mechanisms that underlie synaptic dysfunction and neuropathology in AD.
Article
Full-text available
The only established genetic determinant of non-Mendelian forms of Alzheimer's disease (AD) is the ε4 allele of the apolipoprotein E gene (APOE). Recently, it has been reported that the P86L polymorphism of the calcium homeostasis modulator 1 gene (CALHM1) is associated with the risk of developing AD. In order to independently assess this association, we performed a meta-analysis of 7,873 AD cases and 13,274 controls of Caucasian origin (from a total of 24 centers in Belgium, Finland, France, Italy, Spain, Sweden, the UK, and the USA). Our results indicate that the CALHM1 P86L polymorphism is likely not a genetic determinant of AD but may modulate age of onset by interacting with the effect of the ε4 allele of the APOE gene.
Article
Full-text available
Recent large-scale genetic studies of late-onset Alzheimer's disease have identified risk variants in CALHM1, GAB2, and SORL1. The mechanisms by which these genes might modulate risk are not definitively known. CALHM1 and SORL1 may alter amyloid-β (Aβ) levels and GAB2 may influence phosphorylation of the tau protein. In this study we have analyzed disease associated genetic variants in each of these genes for association with cerebrospinal fluid (CSF) Aβ or tau levels in 602 samples from two independent CSF series. We failed to detect association between CSF Aβ42 levels and single nucleotide polymorphisms in SORL1 despite substantial statistical power to detect association. While we also failed to detect association between variants in GAB2 and CSF tau levels, power to detect this association was limited. Finally, our data suggest that the minor allele of rs2986017, in CALHM1, is marginally associated with CSF Aβ42 levels. This association is consistent with previous reports that this non-synonymous coding substitution results in increased Aβ levels in vitro and provides support for an Aβ-related mechanism for modulating risk for Alzheimer's disease.
Article
J. Neurochem. (2011) 117, 779–796. Regulated intramembrane proteolysis (RIP) controls the communication between cells and the extracellular environment. RIP is essential in the nervous system, but also in other tissues. In the RIP process, a membrane protein typically undergoes two consecutive cleavages. The first one results in the shedding of its ectodomain. The second one occurs within its transmembrane domain, resulting in secretion of a small peptide and the release of the intracellular domain into the cytosol. The proteolytic cleavage fragments act as versatile signaling molecules or are further degraded. An increasing number of membrane proteins undergo RIP. These include growth factors, cytokines, cell adhesion proteins, receptors, viral proteins and signal peptides. A dysregulation of RIP is found in diseases, such as leukemia and Alzheimer’s disease. One of the first RIP substrates discovered was the amyloid precursor protein (APP). RIP processing of APP controls the generation of the amyloid β-peptide, which is believed to cause Alzheimer’s disease. Focusing on APP as the best-studied RIP substrate, this review describes the function and mechanism of the APP RIP proteases with the goal to elucidate cellular mechanisms and common principles of the RIP process in general.
Article
Alzheimer's disease is the largest unmet medical need in neurology. Current drugs improve symptoms, but do not have profound disease-modifying effects. However, in recent years, several approaches aimed at inhibiting disease progression have advanced to clinical trials. Among these, strategies targeting the production and clearance of the amyloid-beta peptide - a cardinal feature of Alzheimer's disease that is thought to be important in disease pathogenesis - are the most advanced. Approaches aimed at modulating the abnormal aggregation of tau filaments (another key feature of the disease), and those targeting metabolic dysfunction, are also being evaluated in the clinic. This article discusses recent progress with each of these strategies, with a focus on anti-amyloid strategies, highlighting the lessons learned and the challenges that remain.